US11753406B2 - Salts of a PD-1/PD-L1 inhibitor - Google Patents

Salts of a PD-1/PD-L1 inhibitor Download PDF

Info

Publication number
US11753406B2
US11753406B2 US16/987,557 US202016987557A US11753406B2 US 11753406 B2 US11753406 B2 US 11753406B2 US 202016987557 A US202016987557 A US 202016987557A US 11753406 B2 US11753406 B2 US 11753406B2
Authority
US
United States
Prior art keywords
salt
compound
cancer
acid salt
xrpd
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US16/987,557
Other languages
English (en)
Other versions
US20210040090A1 (en
Inventor
Zhongjiang Jia
Pingli Liu
David J. Meloni
Yongchun Pan
Yongzhong Wu
Jiacheng Zhou
Qun Li
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Incyte Corp
Original Assignee
Incyte Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Incyte Corp filed Critical Incyte Corp
Priority to US16/987,557 priority Critical patent/US11753406B2/en
Publication of US20210040090A1 publication Critical patent/US20210040090A1/en
Assigned to INCYTE CORPORATION reassignment INCYTE CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WILMINGTON PHARMATECH
Assigned to WILMINGTON PHARMATECH reassignment WILMINGTON PHARMATECH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LI, QUN
Assigned to INCYTE CORPORATION reassignment INCYTE CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JIA, Zhongjiang, LIU, PINGLI, MELONI, DAVID J., PAN, YONGCHUN, WU, YONGZHONG, ZHOU, JIACHENG
Application granted granted Critical
Publication of US11753406B2 publication Critical patent/US11753406B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • This application relates to salt forms of the PD-1/PD-L1 inhibitor (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid, including methods of preparation thereof, where the compound is useful in the treatment of various diseases including infectious diseases and cancer.
  • the immune system plays an important role in controlling and eradicating diseases such as cancer.
  • cancer cells often develop strategies to evade or to suppress the immune system in order to favor their growth.
  • One such mechanism is altering the expression of co-stimulatory and co-inhibitory molecules expressed on immune cells (Postow et al, J. Clinical Oncology 2015, 1-9). Blocking the signaling of an inhibitory immune checkpoint, such as PD-1, has proven to be a promising and effective treatment modality.
  • PD-1 Programmed cell death-1
  • CD279 is a cell surface receptor expressed on activated T cells, natural killer T cells, B cells, and macrophages (Greenwald et al, Annu. Rev. Immunol 2005, 23:515-548; Okazaki and Honjo, Trends Immunol 2006, (4):195-201). It functions as an intrinsic negative feedback system to prevent the activation of T-cells, which in turn reduces autoimmunity and promotes self-tolerance.
  • PD-1 is also known to play a critical role in the suppression of antigen-specific T cell response in diseases like cancer and viral infection (Sharpe et al, Nat Immunol 2007 8, 239-245; Postow et al, J. Clinical Oncol 2015, 1-9).
  • the structure of PD-1 consists of an extracellular immunoglobulin variable-like domain followed by a transmembrane region and an intracellular domain (Parry et al, Mol Cell Biol 2005, 9543-9553).
  • the intracellular domain contains two phosphorylation sites located in an immunoreceptor tyrosine-based inhibitory motif and an immunoreceptor tyrosine-based switch motif, which suggests that PD-1 negatively regulates T cell receptor-mediated signals.
  • PD-1 has two ligands, PD-L1 and PD-L2 (Parry et al, Mol Cell Biol 2005, 9543-9553; Latchman et al, Nat Immunol 2001, 2, 261-268), and they differ in their expression patterns.
  • PD-L1 protein is upregulated on macrophages and dendritic cells in response to lipopolysaccharide and GM-CSF treatment, and on T cells and B cells upon T cell receptor and B cell receptor signaling. PD-L1 is also highly expressed on almost all tumor cells, and the expression is further increased after IFN- ⁇ treatment (Iwai et al, PNAS2002, 99(19):12293-7; Blank et al, Cancer Res 2004, 64(3):1140-5).
  • tumor PD-L1 expression status has been shown to be prognostic in multiple tumor types (Wang et al, Eur J Surg Oncol 2015; Huang et al, Oncol Rep 2015; Sabatier et al, Oncotarget 2015, 6(7): 5449-5464).
  • PD-L2 expression in contrast, is more restricted and is expressed mainly by dendritic cells (Nakae et al, J Immunol 2006, 177:566-73).
  • Ligation of PD-1 with its ligands PD-L1 and PD-L2 on T cells delivers a signal that inhibits IL-2 and IFN- ⁇ production, as well as cell proliferation induced upon T cell receptor activation (Carter et al, Eur J Immunol 2002, 32(3):634-43; Freeman et al, J Exp Med 2000, 192(7):1027-34).
  • the mechanism involves recruitment of SHP-2 or SHP-1 phosphatases to inhibit T cell receptor signaling such as Syk and Lck phosphorylation (Sharpe et al, Nat Immunol 2007, 8, 239-245).
  • Activation of the PD-1 signaling axis also attenuates PKC- ⁇ activation loop phosphorylation, which is necessary for the activation of NF- ⁇ B and AP1 pathways, and for cytokine production such as IL-2, IFN- ⁇ and TNF (Sharpe et al, Nat Immunol 2007, 8, 239-245; Carter et al, Eur J Immunol 2002, 32(3):634-43; Freeman et al, J Exp Med 2000, 192(7):1027-34).
  • PD-1-deficient mice have been shown to develop lupus-like glomerulonephritis and dilated cardiomyopathy (Nishimura et al, Immunity 1999, 11:141-151; Nishimura et al, Science 2001, 291:319-322).
  • LCMV model of chronic infection it has been shown that PD-1/PD-L1 interaction inhibits activation, expansion and acquisition of effector functions of virus-specific CD8 T cells (Barber et al, Nature 2006, 439, 682-7).
  • the present disclosure is directed to salts of (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid.
  • the present disclosure is further directed to the mono-hydrobromic acid salt, the mono-oxalic acid salt, the mono-hydrochloric acid salt, the mono-L-tartaric acid salt, the di-hydrobromic acid salt, the di-oxalic acid salt, the L-tartaric acid salt (1:1.5), the hydrochloric acid salt (1:1.7), the mono-malonic acid salt, the phosphoric acid salt ( ⁇ 1:3), and the phosphoric acid salt ( ⁇ 1:2) of (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid.
  • the present disclosure is further directed to crystalline forms of the salts described herein.
  • the present disclosure is further directed to pharmaceutical compositions comprising a salt or crystalline form described herein, and at least one pharmaceutically acceptable carrier or excipient.
  • the present disclosure is further directed to solid dosage forms comprising the pharmaceutical compositions.
  • the present disclosure is further directed to a method of inhibiting PD-1/PD-L1 interaction comprising administering to a patient the salts and crystalline forms described herein.
  • the present disclosure also provides uses of the salts and crystalline forms described herein in the manufacture of a medicament for use in inhibiting PD-1/PD-L1 interaction.
  • the present disclosure also provides the salts and crystalline forms described herein for use in inhibiting PD-1/PD-L1 interaction.
  • the present disclosure is further directed to treating a disease or disorder associated with inhibition of PD-1/PD-L1 interaction comprising administering to a patient the salts and crystalline forms described herein.
  • the present disclosure also provides uses of the salts and crystalline forms described herein in the manufacture of a medicament for use in treating a disease or disorder associated with inhibition of PD-1/PD-L1 interaction.
  • the present disclosure also provides the salts and crystalline forms described herein for use in treating a disease or disorder associated with inhibition of PD-1/PD-L1 interaction.
  • the present disclosure is further directed to enhancing, stimulating and/or increasing the immune response in a patient comprising administering to a patient the salts and crystalline forms described herein.
  • the present disclosure also provides uses of the salts and crystalline forms described herein in the manufacture of a medicament for use in enhancing, stimulating and/or increasing the immune response in a patient.
  • the present disclosure also provides the salts and crystalline forms described herein for use in enhancing, stimulating and/or increasing the immune response in a patient.
  • the present invention is further directed to processes for preparing the salts and crystalline forms described herein.
  • FIG. 1 shows an XRPD pattern of Compound 1 mono-hydrobromic acid salt.
  • FIG. 2 shows an XRPD pattern of Compound 1 mono-hydrobromic acid salt prepared by an alternative method.
  • FIG. 3 shows an XRPD pattern of Compound 1 mono-oxalic acid salt.
  • FIG. 4 shows a TGA thermogram of Compound 1 mono-oxalic acid salt.
  • FIG. 5 shows an XRPD pattern of Compound 1 mono-hydrochloric acid salt.
  • FIG. 6 shows an XRPD pattern of Compound 1 L-tartaric acid salt (1:1.5).
  • FIG. 7 shows a 1 H NMR of Compound 1 L-tartaric acid salt (1:1.5).
  • FIG. 8 shows an XRPD pattern of two samples of Compound 1 di-hydrobromic acid salt.
  • FIG. 9 shows an XRPD pattern of Compound 1 di-hydrobromic acid salt prepared by an alternative method.
  • FIG. 10 shows an XRPD pattern of Compound 1 di-oxalic acid salt.
  • FIG. 11 shows a DSC thermogram of Compound 1 di-oxalic acid salt.
  • FIG. 12 shows a TGA thermogram of Compound 1 di-oxalic acid salt.
  • FIG. 13 shows an XRPD pattern of Compound 1 L-tartaric acid salt (1:1.7).
  • FIG. 14 shows a 1 H NMR of Compound 1 L-tartaric acid salt (1:1.7).
  • the present disclosure is directed to, inter alia, a salt of (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid (Compound 1), wherein the salt is selected from:
  • the salt of Compound 1 is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid hydrobromic acid salt (Compound 1 hydrobromic acid salt).
  • the Compound 1 hydrobromic acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid mono-hydrobromic acid salt (Compound 1 mono-hydrobromic acid salt).
  • the Compound 1 hydrobromic acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid di-hydrobromic acid salt (Compound 1 di-hydrobromic acid salt).
  • Compound 1 mono-hydrobromic acid salt can be prepared by any suitable method for the preparation of hydrobromic acid addition salts.
  • Compound 1 can be combined with hydrobromic acid (e.g., about 1.0 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • Compound 1 is combined with about 1 to about 2 molar equivalents of hydrobromic acid.
  • Compound 1 is combined with about 1.0 to about 1.5 molar equivalents of hydrobromic acid.
  • Compound 1 is combined with about 1.05 molar equivalents of hydrobromic acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol.
  • Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propanol, or isopropanol. In some embodiments, the solvent contains dichloromethane. In some embodiments, the solvent contains tetrahydrofuran.
  • the solvent is a mixture of isopropyl alcohol, methanol, water, and dichloromethane. In some embodiments, the solvent is a mixture of isopropyl alcohol, water, and tetrahydrofuran.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature. In some embodiments, precipitation is completed within about 12 hours. In some embodiments, precipitation is completed within about 2 hours.
  • the precipitation of Compound 1 mono-hydrobromic acid salt is carried out by filtering the salt from solution.
  • Compound 1 mono-hydrobromic acid salt can be characterized by the X-ray powder diffraction (XRPD) pattern substantially as shown in FIG. 1 . In some embodiments, Compound 1 mono-hydrobromic acid salt can be characterized by the X-ray powder diffraction (XRPD) pattern substantially as shown in FIG. 2 .
  • XRPD X-ray powder diffraction
  • Compound 1 mono-hydrobromic acid salt is amorphous.
  • Compound 1 di-hydrobromic acid salt can be prepared by any suitable method for preparation of hydrobromic acid addition salts.
  • Compound 1 can be combined with hydrobromic acid (e.g., about 2.0 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • Compound 1 is combined with about 2 to about 3 molar equivalents of hydrobromic acid.
  • Compound 1 is combined with about 2.0 to about 2.5 molar equivalents of hydrobromic acid.
  • Compound 1 is combined with about 2.3 molar equivalents of hydrobromic acid.
  • Compound 1 is combined with about 2.05 molar equivalents of hydrobromic acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol.
  • Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propanol, or isopropanol. In some embodiments, the solvent contains methanol. In some embodiments, the solvent contains tetrahydrofuran.
  • the solvent is a mixture of isopropyl alcohol, water and methanol. In some embodiments, the solvent is tetrahydrofuran. In some embodiments, the solvent is a mixture of isopropyl alcohol, water and tetrahydrofuran.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature.
  • the precipitation is completed within about 16 hours.
  • the precipitation is completed within about 12 hours.
  • the precipitation is completed within about 2.5 hours.
  • the precipitation and of the di-hydrobromic acid salt is carried out by filtering the salt from solution.
  • Compound 1 di-hydrobromic acid salt can be characterized by the X-ray powder diffraction (XRPD) pattern substantially as shown in FIG. 8 . In some embodiments, Compound 1 di-hydrobromic acid salt can be characterized by the X-ray powder diffraction (XRPD) pattern substantially as shown in FIG. 9 .
  • Compound 1 di-hydrobromic acid salt is amorphous.
  • the salt of Compound 1 is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid oxalic acid salt (Compound 1 oxalic acid salt).
  • the Compound 1 oxalic acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid mono-oxalic acid salt (Compound 1 mono-oxalic acid salt).
  • the Compound 1 oxalic acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid di-oxalic acid salt (Compound 1 di-oxalic acid salt).
  • Compound 1 mono-oxalic acid salt can be prepared by any suitable method for preparation of oxalic acid addition salts.
  • Compound 1 can be combined with oxalic acid (e.g., about 1.0 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • Compound 1 is combined with about 1 to about 2 molar equivalents of oxalic acid.
  • Compound 1 is combined with about 1.0 to about 1.5 molar equivalents of oxalic acid.
  • Compound 1 is combined with about 1.05 molar equivalents of oxalic acid.
  • Compound 1 is combined with about 1.1 molar equivalents of oxalic acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol. Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propan
  • the solvent is tetrahydrofuran. In some embodiments, the solvent is a mixture of acetone, methanol and dichloromethane.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature.
  • the precipitation is completed within about 2 hours. In some embodiments, the precipitation is completed within about 90 minutes.
  • the precipitation of the mono-oxalic acid salt is carried out by filtering the salt from solution.
  • Compound 1 mono-oxalic acid salt can be characterized by the X-ray powder diffraction (XRPD) pattern substantially as shown in FIG. 3 .
  • Compound 1 mono-oxalic acid salt can be characterized by the thermogravimetric analysis (TGA) spectrum substantially as shown in FIG. 4 .
  • Compound 1 mono-oxalic acid salt is amorphous.
  • Compound 1 di-oxalic acid salt can be prepared by any suitable method for preparation of oxalic acid addition salts.
  • Compound 1 can be combined with oxalic acid (e.g., about 2.0 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • Compound 1 is combined with about 2 to about 3 molar equivalents of oxalic acid.
  • Compound 1 is combined with about 2.0 to about 2.5 molar equivalents of oxalic acid.
  • Compound 1 is combined with about 2.05 molar equivalents of oxalic acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol. Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propan
  • the solvent is tetrahydrofuran.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation and/or crystallization at a practical rate.
  • precipitation and/or crystallization is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation/crystallizing solvent and temperature. In some embodiments, the precipitation and/or crystallization is completed within about 2 hours.
  • the precipitation and/or crystallization of the di-oxalic acid salt is carried out by filtering the salt from solution.
  • Compound 1 di-oxalic acid salt can be characterized by the X-ray powder diffraction (XRPD) pattern substantially as shown in FIG. 10 .
  • XRPD X-ray powder diffraction
  • Compound 1 di-oxalic acid salt has a characteristic X-ray powder diffraction (XRPD) peak at 7.0 ⁇ 0.2 degrees 2-theta. In some embodiments, Compound 1 di-oxalic acid salt has a characteristic X-ray powder diffraction (XRPD) peak at 11.4 ⁇ 0.2 degrees 2-theta. In some embodiments, Compound 1 di-oxalic acid salt has a characteristic X-ray powder diffraction (XRPD) peak at 13.2 ⁇ 0.2 degrees 2-theta. In some embodiments, Compound 1 di-oxalic acid salt has a characteristic X-ray powder diffraction (XRPD) peak at 14.9 ⁇ 0.2 degrees 2-theta. In some embodiments, Compound 1 di-oxalic acid salt has a characteristic X-ray powder diffraction (XRPD) peak at 17.0 ⁇ 0.2 degrees 2-theta.
  • XRPD characteristic X-ray powder diffraction
  • the di-oxalic acid salt of Compound 1 has characteristic X-ray powder diffraction (XRPD) peaks at 7.0 ⁇ 0.2, 11.4 ⁇ 0.2, and 14.9 ⁇ 0.2 degrees 2-theta. In some embodiments, the di-oxalic acid salt of Compound 1 has characteristic X-ray powder diffraction (XRPD) peaks at 7.0 ⁇ 0.2, 11.4 ⁇ 0.2, 14.9 ⁇ 0.2, and 17.0 ⁇ 0.2 degrees 2-theta. In some embodiments, the di-oxalic acid salt of Compound 1 has characteristic X-ray powder diffraction (XRPD) peaks at 7.0 ⁇ 0.2, 11.4 ⁇ 0.2, 13.2 ⁇ 0.2, 14.9 ⁇ 0.2, and 17.0 ⁇ 0.2 degrees 2-theta.
  • XRPD X-ray powder diffraction
  • Compound 1 di-oxalic acid salt has at least one X-ray powder diffraction (XRPD) peak selected from 7.0 ⁇ 0.2, 11.4 ⁇ 0.2, 13.2 ⁇ 0.2, 14.9 ⁇ 0.2, and 17.0 ⁇ 0.2 degrees 2-theta. In some embodiments, Compound 1 di-oxalic acid salt has at least two X-ray powder diffraction (XRPD) peaks selected from 7.0 ⁇ 0.2, 11.4 ⁇ 0.2, 13.2 ⁇ 0.2, 14.9 ⁇ 0.2, and 17.0 ⁇ 0.2 degrees 2-theta.
  • XRPD X-ray powder diffraction
  • Compound 1 di-oxalic acid salt has at least three X-ray powder diffraction (XRPD) peaks selected from 7.0 ⁇ 0.2, 11.4 ⁇ 0.2, 13.2 ⁇ 0.2, 14.9 ⁇ 0.2, and 17.0 ⁇ 0.2 degrees 2-theta. In some embodiments, Compound 1 di-oxalic acid salt has at least four X-ray powder diffraction (XRPD) peaks selected from 7.0 ⁇ 0.2, 11.4 ⁇ 0.2, 13.2 ⁇ 0.2, 14.9 ⁇ 0.2, and 17.0 ⁇ 0.2 degrees 2-theta.
  • XRPD X-ray powder diffraction
  • Compound 1 di-oxalic acid salt exhibits a DSC thermogram having an endothermic peak at a temperature of 235 ⁇ 3° C.
  • the di-oxalic acid salt of Compound 1 has a DSC thermogram substantially as depicted in FIG. 11 .
  • the di-oxalic acid salt of Compound 1 has a TGA thermogram substantially as depicted in FIG. 12 .
  • Compound 1 di-oxalic acid salt has at least one characteristic XRPD peak selected from 7.0 ⁇ 0.2, 11.4 ⁇ 0.2, 13.2 ⁇ 0.2, 14.9 ⁇ 0.2, and 17.0 ⁇ 0.2 degrees 2-theta; and the di-oxalic acid salt of Compound 1 exhibits a DSC thermogram having an endothermic peak at a temperature of 235 ⁇ 3° C.
  • Compound 1 di-oxalic acid salt is amorphous. In some embodiments, Compound 1 di-oxalic acid salt is crystalline. In some embodiments, Compound 1 di-oxalic acid salt is a mixture comprising crystalline and amorphous forms.
  • the salt of Compound 1 is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid hydrochloric acid salt (Compound 1 hydrochloric acid salt).
  • the Compound 1 hydrochloric acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid mono-hydrochloric acid salt (Compound 1 mono-hydrochloric acid salt).
  • the Compound 1 hydrochloric acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid tri-hydrochloric acid salt (Compound 1 tri-hydrochloric acid salt).
  • Compound 1 mono-hydrochloric acid salt can be prepared by any suitable method for preparation of mono-hydrochloric acid addition salts.
  • Compound 1 can be combined with hydrochloric acid (e.g., about 1.0 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • hydrochloric acid e.g., about 1.0 molar eq. or more
  • Compound 1 is combined with about 1 to about 2 molar equivalents of hydrochloric acid.
  • Compound 1 is combined with about 1 to about 1.5 molar equivalents of hydrochloric acid.
  • Compound 1 is combined with about 1.05 molar equivalents of hydrochloric acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol.
  • Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propanol, or isopropanol. In some embodiments, the solvent contains dichloromethane. In some embodiments, the solvent contains methanol.
  • the solvent is a mixture of isopropanol, water, methanol and dichloromethane. In some embodiments, the solvent is a mixture of isopropanol, water and methanol.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature. In some embodiments, precipitation is completed within about 12 hours.
  • the precipitation of the mono-hydrochloric acid salt is carried out by filtering the salt from solution.
  • Compound 1 mono-hydrochloric acid salt can be characterized by the X-ray powder diffraction (XRPD) pattern substantially as shown in FIG. 5 .
  • XRPD X-ray powder diffraction
  • Compound 1 mono-hydrochloric acid salt is amorphous.
  • Compound 1 tri-hydrochloric acid salt can be prepared by any suitable method for preparation of tri-hydrochloric acid addition salts.
  • Compound 1 can be combined with hydrochloric acid (e.g., about 3.0 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • hydrochloric acid e.g., about 3.0 molar eq. or more
  • Compound 1 is combined with about 3 to about 5 molar equivalents of hydrochloric acid.
  • Compound 1 is combined with about 3 to about 4 molar equivalents of hydrochloric acid.
  • Compound 1 is combined with about 4 molar equivalents of hydrochloric acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol.
  • Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains dioxane, dimethylsulfoxide, acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propanol, or isopropanol. In some embodiments, the solvent contains tetrahydrofuran.
  • the solvent is a mixture of dimethylsulfoxide, tetrahydrofuran and dioxane.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature. In some embodiments, precipitation is completed within about 12 hours.
  • the precipitation of the tri-hydrochloric acid salt is carried out by filtering the salt from solution.
  • Compound 1 tri-hydrochloric acid salt is amorphous.
  • the salt of Compound 1 is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid L-tartaric acid salt (Compound 1 L-tartaric acid salt).
  • the Compound 1 L-tartaric acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid L-tartaric acid salt (1:1.5) (Compound 1 L-tartaric acid salt (1:1.5)), wherein the ratio of Compound 1 to L-tartaric acid is about 1 to 1.5.
  • the Compound 1 L-tartaric acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid L-tartaric acid salt (1:1.7) (Compound 1 L-tartaric acid salt (1:1.7)), wherein the ratio of Compound 1 to L-tartaric acid is about 1 to 1.7.
  • Compound 1 L-tartaric acid salt (1:1.5) can be prepared by any suitable method for preparation of L-tartaric acid addition salts.
  • Compound 1 can be combined with L-tartaric acid in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • Compound 1 is combined with about 0.8 to about 2.5 molar equivalents of L-tartaric acid.
  • Compound 1 is combined with about 1.5 to about 2.5 molar equivalents of L-tartaric acid.
  • Compound 1 is combined with about 1.9 to about 2.1 molar equivalents of L-tartaric acid.
  • Compound 1 is combined with about 1.8 to about 2.2 molar equivalents of L-tartaric acid.
  • Compound 1 is combined with about 0.9 to about 1.1 molar equivalents of L-tartaric acid. In certain embodiments, Compound 1 is combined with about 0.8 to about 1.2 molar equivalents of L-tartaric acid. In certain embodiments, Compound 1 is combined with about 0.5 to about 1.5 molar equivalents of L-tartaric acid. In certain embodiments, Compound 1 is combined with about 1.5 to about 2.0 molar equivalents of L-tartaric acid. In certain embodiments, Compound 1 is combined with about 1.5 molar equivalents of L-tartaric acid. In certain embodiments, Compound 1 is combined with about 1.05 molar equivalents of L-tartaric acid. In certain embodiments, Compound 1 is combined with about 2.05 molar equivalents of L-tartaric acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol. Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propan
  • the solvent is tetrahydrofuran.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of at least about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature. In some embodiments, precipitation is completed within about 2 hours.
  • the precipitation of the L-tartaric acid salt (1:1.5), in some embodiments, is carried out by filtering the salt from solution.
  • Compound 1 L-tartaric acid salt (1:1.5) can be characterized by the X-ray powder diffraction (XRPD) pattern substantially as shown in FIG. 6 .
  • Compound 1 L-tartaric acid salt (1:1.5) can be characterized by the 1 H NMR spectrum substantially as depicted in FIG. 7 .
  • Compound 1 L-tartaric acid salt (1:1.5) is amorphous.
  • L-tartaric acid salt (1:1.7) can be prepared by any suitable method for preparation of L-tartaric acid addition salts.
  • Compound 1 can be combined with L-tartaric acid (e.g., about 1.7 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • Compound 1 is combined with about 1.7 to about 3 molar equivalents of L-tartaric acid.
  • Compound 1 is combined with about 1.7 to about 2.5 equivalents of molar L-tartaric acid.
  • Compound 1 is combined with about 2.05 molar equivalents of L-tartaric acid.
  • Compound 1 is combined with about 1.7 molar equivalents of L-tartaric acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol. Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propan
  • the solvent is tetrahydrofuran.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of at least about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature. In some embodiments, precipitation is completed within about 2 hours.
  • the precipitation of the L-tartaric acid salt (1:1.7), in some embodiments, is carried out by filtering the salt from solution.
  • Compound 1 L-tartaric acid salt (1:1.7) can be characterized by the X-ray powder diffraction (XRPD) pattern substantially as shown in FIG. 13 . In some embodiments, Compound 1 L-tartaric acid salt (1:1.7) can be characterized by the 1 H NMR spectrum substantially as depicted in FIG. 14 .
  • XRPD X-ray powder diffraction
  • Compound 1 L-tartaric acid salt (1:1.7) is amorphous.
  • the salt of Compound 1 is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid malonic acid salt (Compound 1 malonic acid salt).
  • the Compound 1 malonic acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid mono-malonic acid salt (Compound 1 mono-malonic acid salt).
  • Compound 1 mono-malonic acid salt can be prepared by any suitable method for preparation of mono-malonic acid addition salts.
  • Compound 1 can be combined with malonic acid (e.g., about 1.0 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • malonic acid e.g., about 1.0 molar eq. or more
  • Compound 1 is combined with about 1 to about 2 molar equivalents of malonic acid.
  • Compound 1 is combined with about 1 to about 1.5 molar equivalents of malonic acid.
  • Compound 1 is combined with about 1.1 molar equivalents of malonic acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol. Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, 1-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propanol
  • the solvent is a mixture of acetone, methanol and dichloromethane.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature.
  • precipitation is completed within about 1 to about 2 hours. In some embodiments, precipitation is completed within about 90 minutes.
  • the precipitation of the mono-malonic acid salt is carried out by filtering the salt from solution.
  • Compound 1 mono-malonic acid salt is amorphous.
  • the salt of Compound 1 is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid phosphoric acid salt (Compound 1 phosphoric acid salt).
  • the Compound 1 phosphoric acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid phosphoric acid salt ( ⁇ 1:2) (Compound 1 phosphoric acid salt ( ⁇ 1:2)), wherein the ratio of Compound 1 to phosphoric acid is about 1 to 2.
  • the Compound 1 phosphoric acid salt is (R)-1-((7-cyano-2-(3′-(3-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-1,7-naphthyridin-8-ylamino)-2,2′-dimethylbiphenyl-3-yl)benzo[d]oxazol-5-yl)methyl)pyrrolidine-3-carboxylic acid phosphoric acid salt ( ⁇ 1:3) (Compound 1 phosphoric acid salt ( ⁇ 1:3) wherein the ratio of Compound 1 to phosphoric acid is about 1 to 3.
  • Compound 1 phosphoric acid salt ( ⁇ 1:2) can be prepared by any suitable method for preparation of phosphoric acid ( ⁇ 1:2) addition salts.
  • Compound 1 can be combined with phosphoric acid (e.g., about 2.0 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • Compound 1 is combined with about 2 to about 3 molar equivalents of phosphoric acid.
  • Compound 1 is combined with about 2 to about 2.5 molar equivalents of phosphoric acid.
  • Compound 1 is combined with about 2 molar equivalents of phosphoric acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol.
  • Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propanol, or isopropanol. In some embodiments, the solvent contains dichloromethane. In some embodiments, the solvent contains acetone.
  • the solvent is a mixture of acetone, methanol and dichloromethane.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature. In some embodiments, the precipitation is completed within about 90 minutes.
  • the precipitation of the phosphoric acid salt ( ⁇ 1:2) is carried out by filtering the salt from solution.
  • Compound 1 phosphoric acid salt ( ⁇ 1:2) is amorphous.
  • Compound 1 phosphoric acid salt ( ⁇ 1:3) can be prepared by any suitable method for preparation of phosphoric acid ( ⁇ 1:3) addition salts.
  • Compound 1 can be combined with phosphoric acid (e.g., about 3.0 molar eq. or more) in a solvent and the resulting salt can be isolated by filtering the salt from solution.
  • Compound 1 is combined with about 3 to about 5 molar equivalents of phosphoric acid.
  • Compound 1 is combined with about 3 to about 4 molar equivalents of phosphoric acid.
  • Compound 1 is combined with about 3 molar equivalents of phosphoric acid.
  • the solvent can contain any solvent or mixture of solvents capable of at least partially dissolving Compound 1.
  • the solvent contains an alcohol. Suitable alcohols include methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, isopropanol (isopropyl alcohol, 2-propanol), 2-methoxyethanol, 1-butanol, 2-butanol, i-butyl alcohol, t-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • the solvent contains acetone, tetrahydrofuran, dichloromethane, methanol, ethanol, 1-propan
  • the solvent is a mixture of acetone, methanol and dichloromethane.
  • the solvent is about room temperature. In some embodiments, the solvent is heated to a temperature of about 50° C. In some embodiments, the temperature is from about 50° C. to about 80° C. In some embodiments, the temperature is from about 40° C. to about 60° C. In some embodiments, the temperature is from about 45° C. to about 55° C. In some embodiments, the temperature is about 45° C., about 50° C., about 55° C., about 60° C., about 65° C., about 70° C., about 75° C. or about 80° C.
  • the solvent is heated to a temperature that can induce precipitation at a practical rate.
  • precipitation is completed within about 12 to about 24 hours, but longer and shorter periods are possible depending on the choice of precipitation solvent and temperature. In some embodiments, the precipitation is completed within about 90 minutes.
  • the precipitation of the phosphoric acid salt ( ⁇ 1:3), in some embodiments, is carried out by filtering the salt from solution.
  • Compound 1 phosphoric acid salt ( ⁇ 1:3) is amorphous.
  • Different forms of the same substance have different bulk properties relating to, for example, hygroscopicity, solubility, stability, and the like.
  • Forms with high melting points often have good thermodynamic stability which is advantageous in prolonging shelf-life drug formulations containing the solid form.
  • Forms with lower melting points often are less thermodynamically stable, but are advantageous in that they have increased water solubility, translating to increased drug bioavailability.
  • Forms that are weakly hygroscopic are desirable for their stability to heat and humidity and are resistant to degradation during long storage.
  • a Compound 1 salt provided herein is crystalline.
  • crystalline or “crystalline form” is meant to refer to a certain lattice configuration of a crystalline substance. Different crystalline forms of the same substance typically have different crystalline lattices (e.g., unit cells) which are attributed to different physical properties that are characteristic of each of the crystalline forms. In some instances, different lattice configurations have different water or solvent content.
  • the different salt forms can be identified by solid state characterization methods such as by X-ray powder diffraction (XRPD). Other characterization methods such as differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), dynamic vapor sorption (DVS), solid state NMR, and the like further help identify the form as well as help determine stability and solvent/water content.
  • XRPD X-ray powder diffraction
  • Other characterization methods such as differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), dynamic vapor sorption (DVS), solid state NMR, and the like further help identify the form as well as help determine stability and solvent/water content.
  • An XRPD pattern of reflections is typically considered a fingerprint of a particular crystalline form. It is well known that the relative intensities of the XRPD peaks can widely vary depending on, inter alia, the sample preparation technique, crystal size distribution, various filters used, the sample mounting procedure, and the particular instrument employed. In some instances, new peaks may be observed or existing peaks may disappear, depending on the type of the instrument or the settings. As used herein, the term “peak” refers to a reflection having a relative height/intensity of at least about 4% of the maximum peak height/intensity. Moreover, instrument variation and other factors can affect the 2-theta values.
  • peak assignments such as those reported herein, can vary by plus or minus about 0.2° (2-theta), and the term “substantially” and “about” as used in the context of XRPD herein is meant to encompass the above-mentioned variations.
  • the term “about” means ⁇ 10%. In some embodiments, the term “about” means ⁇ 5%.
  • the salts described herein are substantially isolated.
  • substantially isolated is meant that the salt is at least partially or substantially separated from the environment in which it was formed or detected.
  • Partial separation can include, for example, a composition enriched in the salts described herein.
  • Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the salts described herein, or salt thereof. Methods for isolating salts are routine in the art.
  • Salts of the invention can also include all isotopes of atoms occurring in the final salts or Compound 1.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • the salts can be found together with other substances such as water and solvents (e.g., hydrates and solvates) or can be isolated.
  • solvents e.g., hydrates and solvates
  • phrases “pharmaceutically acceptable” is employed herein to refer to those salts, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • reaction temperatures will depend on, for example, the melting and boiling points of the reagents and solvent, if present; the thermodynamics of the reaction (e.g., vigorously exothermic reactions may need to be carried out at reduced temperatures); and the kinetics of the reaction (e.g., a high activation energy barrier may need elevated temperatures).
  • ambient temperature and “room temperature” or “rt” as used herein, are understood in the art, and refer generally to a temperature, e.g., a reaction temperature, that is about the temperature of the room in which the salt forming reaction is carried out, for example, a temperature from about 20° C. to about 30° C.
  • the salt forming reactions described herein can be carried out in air or under an inert atmosphere.
  • reactions containing reagents or products that are substantially reactive with air can be carried out using air-sensitive synthetic techniques that are well known to the skilled artisan.
  • Salts of the present disclosure can inhibit the activity of PD-1/PD-L1 protein/protein interaction and, thus, are useful in treating diseases and disorders associated with activity of PD-1 and the diseases and disorders associated with PD-L1 including its interaction with other proteins such as PD-1 and B7-1 (CD80).
  • the salts of the present disclosure are useful for therapeutic administration to enhance, stimulate and/or increase immunity in cancer, chronic infection or sepsis, including enhancement of response to vaccination.
  • the present disclosure provides a method for inhibiting the PD-1/PD-L1 protein/protein interaction.
  • the method includes administering to an individual or a patient a salt of Compound 1, or of a salt as recited in any of the claims and described herein, or a pharmaceutically acceptable salt or a stereoisomer thereof.
  • the salts of the present disclosure can be used alone, in combination with other agents or therapies or as an adjuvant or neoadjuvant for the treatment of diseases or disorders, including cancer or infection diseases.
  • any of the salts of the disclosure including any of the embodiments thereof, may be used.
  • the salts of the present disclosure inhibit the PD-1/PD-L1 protein/protein interaction, resulting in a PD-1 pathway blockade.
  • the blockade of PD-1 can enhance the immune response to cancerous cells and infectious diseases in mammals, including humans.
  • the present disclosure provides treatment of an individual or a patient in vivo using a salt of Compound 1 such that growth of cancerous tumors is inhibited.
  • a salt of Compound 1, or a salt as recited in any of the claims and described herein can be used to inhibit the growth of cancerous tumors.
  • a salt of Compound 1, or a salt as recited in any of the claims and described herein can be used in conjunction with other agents or standard cancer treatments, as described below.
  • the present disclosure provides a method for inhibiting growth of tumor cells in vitro.
  • the method includes contacting the tumor cells in vitro with a salt of Compound 1, or of a salt as recited in any of the claims and described herein.
  • the present disclosure provides a method for inhibiting growth of tumor cells in an individual or a patient. The method includes administering to the individual or patient in need thereof a therapeutically effective amount of a salt of Compound 1, or of a salt as recited in any of the claims and described herein.
  • a method for treating cancer includes administering to a patient in need thereof, a therapeutically effective amount of a salt of Compound 1, or a salt as recited in any of the claims and described herein.
  • cancers include those whose growth may be inhibited using salts of the disclosure and cancers typically responsive to immunotherapy.
  • the present disclosure provides a method of enhancing, stimulating and/or increasing the immune response in a patient.
  • the method includes administering to the patient in need thereof a therapeutically effective amount of a salt of Compound 1, or a salt or composition as recited in any of the claims and described herein.
  • cancers that are treatable using the salts of the present disclosure include, but are not limited to, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, testicular cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, endometrial cancer, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, non-Hodgkin's lymphoma, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, chronic or acute leukemias including acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic le
  • cancers treatable with salts of the present disclosure include melanoma (e.g., metastatic malignant melanoma, cutaneous melanoma), renal cancer (e.g., clear cell carcinoma), prostate cancer (e.g., hormone refractory prostate adenocarcinoma), breast cancer (e.g., breast invasive carcinoma), colon cancer, lung cancer (e.g., non-small cell lung cancer and small cell lung cancer), squamous cell head and neck cancer (e.g., squamous cell carcinoma of the head and neck), urothelial cancer (e.g., bladder cancer, nonmuscle invasive bladder cancer (NMIBC)) and cancers with high microsatellite instability (MSI high ). Additionally, the disclosure includes refractory or recurrent malignancies whose growth may be inhibited using the salts of the disclosure.
  • melanoma e.g., metastatic malignant melanoma, cutaneous melanoma
  • renal cancer e
  • cancers that are treatable using the salts of the present disclosure include, but are not limited to, solid tumors (e.g., prostate cancer, colon cancer, esophageal cancer, endometrial cancer, ovarian cancer, uterine cancer, renal cancer, hepatic cancer, pancreatic cancer, gastric cancer, breast cancer, lung cancer, cancers of the head and neck, thyroid cancer, glioblastoma, sarcoma, bladder cancer, etc.), hematological cancers (e.g., lymphoma, leukemia such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), DLBCL, mantle cell lymphoma, Non-Hodgkin lymphoma (including relapsed or refractory NHL and recurrent follicular), Hodgkin lymphoma or multiple myeloma) and
  • cancers that are treatable using the salts of the present disclosure include, but are not limited to, cholangiocarcinoma, bile duct cancer, biliary tract cancer, triple negative breast cancer, rhabdomyosarcoma, small cell lung cancer, leiomyosarcoma, hepatocellular carcinoma, Ewing's sarcoma, brain cancer, brain tumor, astrocytoma, neuroblastoma, neurofibroma, basal cell carcinoma, chondrosarcoma, epithelioid sarcoma, eye cancer, Fallopian tube cancer, gastrointestinal cancer, gastrointestinal stromal tumors, hairy cell leukemia, intestinal cancer, islet cell cancer, oral cancer, mouth cancer, throat cancer, laryngeal cancer, lip cancer, mesothelioma, neck cancer, nasal cavity cancer, ocular cancer, ocular melanoma, pelvic cancer, rectal cancer, renal cell carcinoma, salivary gland cancer, sinus cancer, spinal cancer, tongue
  • the salts of the present disclosure can be used to treat sickle cell disease and sickle cell anemia.
  • diseases and indications that are treatable using the salts of the present disclosure include, but are not limited to hematological cancers, sarcomas, lung cancers, gastrointestinal cancers, genitourinary tract cancers, liver cancers, bone cancers, nervous system cancers, gynecological cancers, and skin cancers.
  • Exemplary hematological cancers include lymphomas and leukemias such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma, Non-Hodgkin lymphoma (including relapsed or refractory NHL and recurrent follicular), Hodgkin lymphoma, myeloproliferative diseases (e.g., primary myelofibrosis (PMF), polycythemia vera (PV), and essential thrombocytosis (ET)), myelodysplasia syndrome (MDS), T-cell acute lymphoblastic lymphoma (T-ALL) and multiple myeloma (MM).
  • ALL acute lymphoblastic leukemia
  • AML acute mye
  • Exemplary sarcomas include chondrosarcoma, Ewing's sarcoma, osteosarcoma, rhabdomyosarcoma, angiosarcoma, fibrosarcoma, liposarcoma, myxoma, rhabdomyoma, rhabdosarcoma, fibroma, lipoma, harmatoma, and teratoma.
  • Exemplary lung cancers include non-small cell lung cancer (NSCLC) (e.g., squamous cell NSCLC), small cell lung cancer, bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, chondromatous hamartoma, and mesothelioma.
  • NSCLC non-small cell lung cancer
  • small cell lung cancer e.g., squamous cell NSCLC
  • bronchogenic carcinoma squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma
  • alveolar (bronchiolar) carcinoma bronchial adenoma
  • chondromatous hamartoma chondromatous hamartoma
  • mesothelioma mesothelioma
  • Exemplary gastrointestinal cancers include cancers of the esophagus (carcinoma, squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma, adenocarcinoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), and colorectal cancer (e.g., colorectal adeno
  • Exemplary genitourinary tract cancers include cancers of the kidney (adenocarcinoma, Wilm's tumor [nephroblastoma]), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), and testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma).
  • the cancer is a urological cancer (e.g., papillary kidney carcinoma, testicular germ cell cancer, chromophobe renal cell carcinoma, clear cell renal carcinoma, or prostate adenocarcinoma).
  • liver cancers include hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, and hemangioma.
  • Exemplary bone cancers include, for example, osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma, and giant cell tumors.
  • osteogenic sarcoma osteosarcoma
  • fibrosarcoma malignant fibrous histiocytoma
  • chondrosarcoma chondrosarcoma
  • Ewing's sarcoma malignant lymphoma
  • multiple myeloma malignant giant cell tumor chordoma
  • osteochronfroma osteocar
  • Exemplary nervous system cancers include cancers of the skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, meduoblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma, glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), and spinal cord (neurofibroma, meningioma, glioma, sarcoma), as well as neuroblastoma and Lhermitte-Duclos disease.
  • skull osteoma, hemangioma, granuloma, xanthoma, osteitis de
  • Exemplary gynecological cancers include cancers of the uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma (serous cystadenocarcinoma, serous adenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), and fallopian tubes (carcinoma).
  • endometrial carcinoma endometrial carcinoma
  • cervix cervical carcinoma, pre-tum
  • Exemplary skin cancers include melanoma, basal cell carcinoma, squamous cell carcinoma (e.g., cutaneous squamous cell carcinoma), Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, and keloids.
  • diseases and indications that are treatable using the salts of the present disclosure include, but are not limited to, sickle cell disease (e.g., sickle cell anemia), triple-negative breast cancer (TNBC), myelodysplastic syndromes, testicular cancer, bile duct cancer, esophageal cancer, and urothelial carcinoma.
  • PD-1 pathway blockade with salts of the present disclosure can also be used for treating infections such as viral, bacteria, fungus and parasite infections.
  • the present disclosure provides a method for treating infections such as viral infections. The method includes administering to a patient in need thereof, a therapeutically effective amount of a salt of Compound 1, or a salt as recited in any of the claims and described herein.
  • viruses causing infections treatable by methods of the present disclosure include, but are not limited to, human immunodeficiency virus, human papillomavirus, influenza, hepatitis A, B, C or D viruses, adenovirus, poxvirus, herpes simplex viruses, human cytomegalovirus, severe acute respiratory syndrome virus, ebola virus, and measles virus.
  • viruses causing infections treatable by methods of the present disclosure include, but are not limited to, hepatitis (A, B, or C), herpes virus (e.g., VZV, HSV-1, HAV-6, HSV-II, and CMV, Epstein Barr virus), adenovirus, influenza virus, flaviviruses, echovirus, rhinovirus, coxsackie virus, coronavirus, respiratory syncytial virus, mumps virus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, dengue virus, papillomavirus, molluscum virus, poliovirus, rabies virus, JC virus, tuberculosis and arboviral encephalitis virus.
  • herpes virus e.g., VZV, HSV-1, HAV-6, HSV-II, and CMV, Epstein Barr virus
  • adenovirus e.g., adenovirus
  • the present disclosure provides a method for treating bacterial infections.
  • the method includes administering to a patient in need thereof, a therapeutically effective amount of a salt of Compound 1, or a salt as recited in any of the claims and described herein.
  • pathogenic bacteria causing infections treatable by methods of the disclosure include chlamydia , rickettsial bacteria, mycobacteria, staphylococci, streptococci, pneumococci, meningococci and conococci, klebsiella, proteus, serratia, pseudomonas, legionella, diphtheria, salmonella , bacilli, cholera, tetanus, botulism, anthrax, plague, leptospirosis, and Lyme's disease bacteria.
  • the present disclosure provides a method for treating fungus infections.
  • the method includes administering to a patient in need thereof, a therapeutically effective amount of a salt of Compound 1, or a salt as recited in any of the claims and described herein.
  • pathogenic fungi causing infections treatable by methods of the disclosure include Candida ( albicans, krusei, glabrata, tropicalis , etc.), Cryptococcus neoformans, Aspergillus ( fumigatus, niger , etc.), Genus Mucorales ( mucor, absidia, rhizophus ), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum.
  • the present disclosure provides a method for treating parasite infections.
  • the method includes administering to a patient in need thereof, a therapeutically effective amount of a salt of Compound 1, or a salt as recited in any of the claims and described herein.
  • pathogenic parasites causing infections treatable by methods of the disclosure include Entamoeba histolytica, Balantidium coli, Naegleriafowleri, Acanthamoeba sp., Giardia lambia, Cryptosporidium sp., Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondi , and Nippostrongylus brasiliensis.
  • the present disclosure provides a method for treating neurodegenerative diseases or disorders.
  • the method includes administering to a patient in need thereof, a therapeutically effective amount of a salt of Compound 1, or a salt as recited in any of the claims and described herein.
  • neurodegenerative diseases or disorders include Alzheimer's disease, Parkinson's disease, Huntington's disease, prion disease, Motor neurone diseases, Spinocerebellar ataxia and Spinal muscular atrophy.
  • salts of Compound 1, or any of the embodiments thereof may possess satisfactory pharmacological profile and promising biopharmaceutical properties, such as toxicological profile, metabolism and pharmacokinetic properties, solubility, and permeability. It will be understood that determination of appropriate biopharmaceutical properties is within the knowledge of a person skilled in the art, e.g., determination of cytotoxicity in cells or inhibition of certain targets or channels to determine potential toxicity.
  • mice refer to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • terapéuticaally effective amount refers to the amount of active salt that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
  • treating refers to one or more of (1) inhibiting the disease; e.g., inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (2) ameliorating the disease; e.g., ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease.
  • the salts of the invention are useful in preventing or reducing the risk of developing any of the diseases referred to herein; e.g., preventing or reducing the risk of developing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease.
  • Salts of the present disclosure can be used in combination with one or more immune checkpoint inhibitors for the treatment of diseases, such as cancer or infections.
  • immune checkpoint inhibitors include inhibitors against immune checkpoint molecules such as CBL-B, CD20, CD122, CD96, CD73, CD47, CSF1R, JAK, PI3K delta, PI3K gamma, TAM, arginase, HPK1, A2AR, B7-H3, B7-H4, BTLA, CTLA-4, LAG3, TIM3, TIGIT, CD112R, VISTA, PD-1, PD-L1 and PD-L2.
  • immune checkpoint inhibitors include inhibitors against immune checkpoint molecules such as CBL-B, CD20, CD122, CD96, CD73, CD47, CSF1R, JAK, PI3K delta, PI3K gamma, TAM, arginase, HPK1, A2AR, B7-H3, B7-H4, BTLA, CTLA-4, LAG
  • the immune checkpoint molecule is a stimulatory checkpoint molecule selected from CD27, CD28, CD40, ICOS, OX40, GITR and CD137 (4-1BB).
  • the immune checkpoint molecule is an inhibitory checkpoint molecule selected from A2AR, B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR, LAG3, PD-1, TIM3, TIGIT, and VISTA.
  • the salts provided herein can be used in combination with one or more agents selected from KIR inhibitors, TIGIT inhibitors, LAIR1 inhibitors, CD160 inhibitors, 2B4 inhibitors and TGF beta inhibitors.
  • the salts provided herein can be used in combination with one or more agonists of immune checkpoint molecules, e.g., OX40, CD27, GITR, and CD137 (also known as 4-1B).
  • one or more agonists of immune checkpoint molecules e.g., OX40, CD27, GITR, and CD137 (also known as 4-1B).
  • the agonist of an immune checkpoint molecule is an agonist of OX40, CD27, CD28, GITR, ICOS, CD40, TLR7/8, and CD137 (also known as 4-1B).
  • the agonist of CD137 is urelumab. In some embodiments, the agonist of CD137 is utomilumab.
  • the agonist of an immune checkpoint molecule is an agonist of CD40.
  • the agonist of CD40 is CP-870893, ADC-1013, CDX-1140, SEA-CD40, RO7009789, JNJ-64457107, APX-005M, or Chi Lob 7/4.
  • the agonist of an immune checkpoint molecule is an agonist of ICOS.
  • the agonist of ICOS is GSK-3359609, JTX-2011, or MEDI-570.
  • the agonist of an immune checkpoint molecule is an agonist of CD28. In some embodiments, the agonist of CD28 is theralizumab.
  • the agonist of an immune checkpoint molecule is an agonist of CD27. In some embodiments, the agonist of CD27 is varlilumab.
  • the agonist of an immune checkpoint molecule is an agonist of TLR7/8. In some embodiments, the agonist of TLR7/8 is MEDI9197.
  • the inhibitor of an immune checkpoint molecule is anti-PD1 antibody, anti-PD-L1 antibody, or anti-CTLA-4 antibody.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1, e.g., an anti-PD-1 monoclonal antibody.
  • the anti-PD-1 monoclonal antibody is nivolumab, pembrolizumab (also known as MK-3475), pidilizumab, cemiplimab, spartalizumab, camrelizumab, cetrelimab, toripalimab, sintilimab, SHR-1210, PDR001, MGA012, PDR001, AB122, AMP-224, JTX-4014, BGB-108, BCD-100, BAT1306, LZM009, AK105, HLX10, or TSR-042.
  • the anti-PD-1 monoclonal antibody is nivolumab or pembrolizumab. In some embodiments, the anti-PD1 antibody is pembrolizumab. In some embodiments, the anti-PD-1 monoclonal antibody is MGA012. In some embodiments, the anti-PD1 antibody is SHR-1210.
  • Other anti-cancer agent(s) include antibody therapeutics such as 4-1BB (e.g. urelumab, utomilumab).
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-L1, e.g., an anti-PD-L1 monoclonal antibody.
  • the anti-PD-L1 monoclonal antibody is BMS-935559, MEDI4736, MPDL3280A (also known as RG7446), durvalumab (Imfinzi®), atezolizumab (Tecentriq®), Avelumab (Bavencio®), MSB0010718C, tislelizumab, FAZ053, KN035, CS1001, SHR-1316, CBT-502, A167, STI-A101, CK-301, BGB-A333, MSB-2311, HLX20, or LY3300054.
  • the anti-PD-L1 monoclonal antibody is MPDL3280A or MEDI4736.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1 and PD-L1, e.g., an anti-PD-1/PD-L1 bispecific antibody.
  • the anti-PD-1/PD-L1 bispecific antibody is MCLA-136.
  • the inhibitor is MCLA-145.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA-4, e.g., an anti-CTLA-4 antibody.
  • the anti-CTLA-4 antibody is ipilimumab, tremelimumab, AGEN1884, or CP-675,206.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1 and CTLA-4, e.g., an anti-PD-1/CTLA-4 bispecific antibody.
  • the anti-PD-1/CTLA-4 antibody is AK104.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of LAG3, e.g., an anti-LAG3 antibody.
  • the anti-LAG3 antibody is BMS-986016, LAG525, INCAGN2385, or eftilagimod alpha (IMP321).
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CD73. In some embodiments, the inhibitor of CD73 is oleclumab. In some embodiments, the inhibitor of CD73 is MEDI9447.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of TIGIT. In some embodiments, the inhibitor of TIGIT is OMP-31M32.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of VISTA.
  • the inhibitor of VISTA is JNJ-61610588 or CA-170.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of B7-H3.
  • the inhibitor of B7-H3 is enoblituzumab, MGD009, or 8H9.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of KIR.
  • the inhibitor of KIR is lirilumab or IPH4102.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of A2aR. In some embodiments, the inhibitor of A2aR is CPI-444.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of TGF-beta.
  • the inhibitor of TGF-beta is trabedersen, galusertinib, or M7824.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PI3K-gamma. In some embodiments, the inhibitor of PI3K-gamma is IPI-549.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CD47.
  • the inhibitor of CD47 is Hu5F9-G4 or TTI-621.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CD70.
  • the inhibitor of CD70 is cusatuzumab or BMS-936561.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of TIM3, e.g., an anti-TIM3 antibody.
  • the anti-TIM3 antibody is INCAGN2390, MBG453, or TSR-022.
  • the agonist of an immune checkpoint molecule is an agonist of GITR, e.g., an anti-GITR antibody.
  • the agonist is TRX518, MK-4166, INCAGN1876, MK-1248, AMG228, BMS-986156, GWN323, MEDI1873, or MEDI6469.
  • the agonist of an immune checkpoint molecule is an agonist of OX40, e.g., OX40 agonist antibody or OX40L fusion protein.
  • OX40 e.g., OX40 agonist antibody or OX40L fusion protein.
  • the anti-OX40 antibody is MEDI0562, MOXR-0916, PF-04518600, GSK3174998, BMS-986178, or 9B12.
  • the OX40L fusion protein is MEDI6383.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CD20, e.g., an anti-CD20 antibody.
  • the anti-CD20 antibody is obinutuzumab or rituximab.
  • the salts of the present disclosure can be used in combination with bispecific antibodies.
  • one of the domains of the bispecific antibody targets PD-1, PD-L1, CTLA-4, GITR, OX40, TIM3, LAG3, CD137, ICOS, CD3 or TGF ⁇ receptor.
  • the bispecific antibody binds to PD-1 and PD-L1.
  • the bispecific antibody that binds to PD-1 and PD-L1 is MCLA-136.
  • the bispecific antibody binds to PD-L1 and CTLA-4.
  • the bispecific antibody that binds to PD-L1 and CTLA-4 is AK104.
  • the salts of the disclosure can be used in combination with one or more metabolic enzyme inhibitors.
  • the metabolic enzyme inhibitor is an inhibitor of IDO1, TDO, or arginase.
  • IDO1 inhibitors include epacadostat, NLG919, BMS-986205, PF-06840003, IOM2983, RG-70099 and LY338196.
  • the additional compounds, inhibitors, agents, etc. can be combined with the present salt in a single or continuous dosage form, or they can be administered simultaneously or sequentially as separate dosage forms.
  • Cancer cell growth and survival can be impacted by dysfunction in multiple biological pathways.
  • Targeting more than one signaling pathway (or more than one biological molecule involved in a given signaling pathway) may reduce the likelihood of drug-resistance arising in a cell population, or reduce the toxicity of treatment.
  • the salts of the present disclosure can be used in combination with one or more other therapies for the treatment of diseases, such as cancer or infections.
  • diseases and indications treatable with combination therapies include those as described herein.
  • cancers include solid tumors and non-solid tumors, such as liquid tumors, blood cancers.
  • infections include viral infections, bacterial infections, fungus infections or parasite infections.
  • the salts of the present disclosure can be combined with one or more inhibitors of the following kinases for the treatment of cancer: Akt1, Akt2, Akt3, BCL2, CDK, TGF- ⁇ R, PKA, PKG, PKC, CaM-kinase, phosphorylase kinase, MEKK, ERK, MAPK, mTOR, EGFR, HER2, HER3, HER4, INS-R, IDH2, IGF-1R, IR-R, PDGF ⁇ R, PDGF ⁇ R, PI3K (alpha, beta, gamma, delta, and multiple or selective), CSF1R, KIT, FLK-II, KDR/FLK-1, FLK-4, fit-1, FGFR1, FGFR2, FGFR3, FGFR4, c-Met, PARP, Ron, Sea, TRKA, TRKB, TRKC, TAM kinases (Axl, Mer, Tyro3), FLT3, VEGFR/Flt2, Flt4,
  • the salts of the present disclosure can be combined with one or more of the following inhibitors for the treatment of cancer or infections.
  • inhibitors that can be combined with the salts of the present disclosure for treatment of cancer and infections include an FGFR inhibitor (FGFR1, FGFR2, FGFR3 or FGFR4, e.g., pemigatinib (INCY54828), INCB62079), an EGFR (also known as ErB-1 or HER-1) inhibitor (e.g., erlotinib, gefitinib, vandetanib, orsimertinib, cetuximab, necitumumab, or panitumumab), a VEGFR inhibitor or pathway blocker (e.g., bevacizumab, pazopanib, sunitinib, sorafenib, axitinib, regorafenib, ponatinib, cabozantinib, vandetani
  • FGFR inhibitor
  • the salts of the present disclosure can be combined with a TLR7 agonist (e.g., imiquimod).
  • a TLR7 agonist e.g., imiquimod
  • the salts of the present disclosure can further be used in combination with other methods of treating cancers, for example by chemotherapy, irradiation therapy, tumor-targeted therapy, adjuvant therapy, immunotherapy or surgery.
  • immunotherapy include cytokine treatment (e.g., interferons, GM-CSF, G-CSF, IL-2), CRS-207 immunotherapy, cancer vaccine, monoclonal antibody, bispecific or multi-specific antibody, antibody drug conjugate, adoptive T cell transfer, Toll receptor agonists, STING agonists, RIG-I agonists, oncolytic virotherapy and immunomodulating small molecules, including thalidomide or JAK1/2 inhibitor, PI3K ⁇ inhibitor and the like.
  • the salts can be administered in combination with one or more anti-cancer drugs, such as a chemotherapeutic agent.
  • chemotherapeutics include any of abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, anastrozole, arsenic trioxide, asparaginase, azacitidine, bevacizumab, bexarotene, baricitinib, bleomycin, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, dalteparin sodium, dasatinib, daunorubicin, decitabine,
  • anti-cancer agent(s) include antibody therapeutics such as trastuzumab (Herceptin), antibodies to costimulatory molecules such as CTLA-4 (e.g., ipilimumab), 4-1BB (e.g., urelumab, utomilumab), antibodies to PD-1 and PD-L1, or antibodies to cytokines (IL-10, TGF- ⁇ , etc.).
  • trastuzumab Herceptin
  • CTLA-4 e.g., ipilimumab
  • 4-1BB e.g., urelumab, utomilumab
  • PD-1 and PD-L1 antibodies to cytokines (IL-10, TGF- ⁇ , etc.
  • antibodies to PD-1 and/or PD-L1 that can be combined with salts of the present disclosure for the treatment of cancer or infections such as viral, bacteria, fungus and parasite infections include, but are not limited to nivolumab, pembrolizumab, atezolizumab, durvalumab, avelumab and SHR-1210.
  • the salts of the present disclosure can further be used in combination with one or more anti-inflammatory agents, steroids, immunosuppressants or therapeutic antibodies.
  • the salts of Compound 1, or a salt as recited in any of the claims and described herein, can be combined with another immunogenic agent, such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines.
  • tumor vaccines include peptides of melanoma antigens, such as peptides of gp100, MAGE antigens, Trp-2, MARTI and/or tyrosinase, or tumor cells transfected to express the cytokine GM-CSF.
  • the salts of Compound 1, or a salt as recited in any of the claims and described herein, can be used in combination with a vaccination protocol for the treatment of cancer.
  • the tumor cells are transduced to express GM-CSF.
  • tumor vaccines include the proteins from viruses implicated in human cancers such as Human Papilloma Viruses (HPV), Hepatitis Viruses (HBV and HCV) and Kaposi's Herpes Sarcoma Virus (KHSV).
  • the salts of the present disclosure can be used in combination with tumor specific antigen such as heat shock proteins isolated from tumor tissue itself.
  • the salts of Compound 1, or a salt as recited in any of the claims and described herein can be combined with dendritic cells immunization to activate potent anti-tumor responses.
  • the salts of the present disclosure can be used in combination with bispecific macrocyclic peptides that target Fe alpha or Fe gamma receptor-expressing effectors cells to tumor cells.
  • the salts of the present disclosure can also be combined with macrocyclic peptides that activate host immune responsiveness.
  • the salts of the present disclosure can be used in combination with bone marrow transplant for the treatment of a variety of tumors of hematopoietic origin.
  • the salts of Compound 1, or a salt as recited in any of the claims and described herein, can be used in combination with vaccines, to stimulate the immune response to pathogens, toxins, and self antigens.
  • pathogens for which this therapeutic approach may be particularly useful include pathogens for which there is currently no effective vaccine, or pathogens for which conventional vaccines are less than completely effective. These include, but are not limited to, HIV, Hepatitis (A, B, & C), Influenza, Herpes, Giardia, Malaria, Leishmania, Staphylococcus aureus, Pseudomonas Aeruginosa.
  • Viruses causing infections treatable by methods of the present disclosure include, but are not limited to human papillomavirus, influenza, hepatitis A, B, C or D viruses, adenovirus, poxvirus, herpes simplex viruses, human cytomegalovirus, severe acute respiratory syndrome virus, ebola virus, measles virus, herpes virus (e.g., VZV, HSV-1, HAV-6, HSV-II, and CMV, Epstein Barr virus), flaviviruses, echovirus, rhinovirus, coxsackie virus, coronavirus, respiratory syncytial virus, mumpsvirus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, dengue virus, papillomavirus, molluscum virus, poliovirus, rabies virus, JC virus and arboviral encephalitis virus.
  • human papillomavirus influenza, hepatitis A
  • Pathogenic bacteria causing infections treatable by methods of the disclosure include, but are not limited to, chlamydia , rickettsial bacteria, mycobacteria, staphylococci, streptococci, pneumococci, meningococci and conococci, klebsiella, proteus, serratia, pseudomonas, legionella, diphtheria, salmonella , bacilli, cholera, tetanus, botulism, anthrax, plague, leptospirosis, and Lyme's disease bacteria.
  • Pathogenic fungi causing infections treatable by methods of the disclosure include, but are not limited to, Candida ( albicans, krusei, glabrata, tropicalis , etc.), Cryptococcus neoformans, Aspergillus ( fumigatus, niger , etc.), Genus Mucorales ( mucor, absidia, rhizophus ), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum.
  • Candida albicans, krusei, glabrata, tropicalis , etc.
  • Cryptococcus neoformans Aspergillus ( fumigatus, niger , etc.)
  • Genus Mucorales mucor, absidia, rhizophus
  • Sporothrix schenkii Blastomyces dermatitidis
  • Pathogenic parasites causing infections treatable by methods of the disclosure include, but are not limited to, Entamoeba histolytica, Balantidium coli, Naegleriafowleri, Acanthamoeba sp., Giardia lambia, Cryptosporidium sp., Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondi , and Nippostrongylus brasiliensis.
  • more than one pharmaceutical agent When more than one pharmaceutical agent is administered to a patient, they can be administered simultaneously, separately, sequentially, or in combination (e.g., for more than two agents).
  • the salts of the present disclosure can be administered in the form of pharmaceutical compositions.
  • the present disclosure provides a composition comprising a salt of Compound 1, or a salt as recited in any of the claims and described herein, or any of the embodiments thereof, and at least one pharmaceutically acceptable carrier or excipient.
  • These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is indicated and upon the area to be treated.
  • Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral.
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration.
  • Parenteral administration can be in the form of a single bolus dose, or may be, e.g., by a continuous perfusion pump.
  • compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • compositions which contain, as the active ingredient, the salt of the present disclosure, in combination with one or more pharmaceutically acceptable carriers or excipients.
  • the composition is suitable for topical administration.
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, e.g., a capsule, sachet, paper, or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, e.g., up to 10% by weight of the active salt, soft and hard gelatin capsules, suppositories, sterile injectable solutions and sterile packaged powders.
  • the active salt can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active salt is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active salt is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
  • the salts of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types.
  • Finely divided (nanoparticulate) preparations of the salts of the invention can be prepared by processes known in the art see, e.g., WO 2002/000196.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • the compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • the pharmaceutical composition comprises silicified microcrystalline cellulose (SMCC) and at least one salt described herein.
  • SMCC silicified microcrystalline cellulose
  • the silicified microcrystalline cellulose comprises about 98% microcrystalline cellulose and about 2% silicon dioxide w/w.
  • the composition is a sustained release composition comprising at least one salt described herein, and at least one pharmaceutically acceptable carrier or excipient.
  • the composition comprises at least one salt described herein, and at least one component selected from microcrystalline cellulose, lactose monohydrate, hydroxypropyl methylcellulose and polyethylene oxide.
  • the composition comprises at least one salt described herein, and microcrystalline cellulose, lactose monohydrate and hydroxypropyl methylcellulose.
  • the composition comprises at least one salt described herein, and microcrystalline cellulose, lactose monohydrate and polyethylene oxide.
  • the composition further comprises magnesium stearate or silicon dioxide.
  • the microcrystalline cellulose is Avicel PH102TM.
  • the lactose monohydrate is Fast-flo 316TM.
  • the hydroxypropyl methylcellulose is hydroxypropyl methylcellulose 2208 K4M (e.g., Methocel K4 M PremierTM) and/or hydroxypropyl methylcellulose 2208 K100LV (e.g., Methocel KOOLVTM).
  • the polyethylene oxide is polyethylene oxide WSR 1105 (e.g., Polyox WSR 1105TM).
  • a wet granulation process is used to produce the composition. In some embodiments, a dry granulation process is used to produce the composition.
  • compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1,000 mg (1 g), more usually about 100 mg to about 500 mg, of the active ingredient. In some embodiments, each dosage contains about 10 mg of the active ingredient. In some embodiments, each dosage contains about 50 mg of the active ingredient. In some embodiments, each dosage contains about 25 mg of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the components used to formulate the pharmaceutical compositions are of high purity and are substantially free of potentially harmful contaminants (e.g., at least National Food grade, generally at least analytical grade, and more typically at least pharmaceutical grade).
  • the composition is preferably manufactured or formulated under Good Manufacturing Practice standards as defined in the applicable regulations of the U.S. Food and Drug Administration.
  • suitable formulations may be sterile and/or substantially isotonic and/or in full compliance with all Good Manufacturing Practice regulations of the U.S. Food and Drug Administration.
  • the active salt may be effective over a wide dosage range and is generally administered in a therapeutically effective amount. It will be understood, however, that the amount of the salt actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual salt administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms and the like.
  • the therapeutic dosage of a salt of the present invention can vary according to, e.g., the particular use for which the treatment is made, the manner of administration of the salt, the health and condition of the patient, and the judgment of the prescribing physician.
  • the proportion or concentration of a salt of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration.
  • the salts of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the salt for parenteral administration. Some typical dose ranges are from about 1 ⁇ g/kg to about 1 g/kg of body weight per day.
  • the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the salt selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a salt of the present invention.
  • a solid preformulation composition containing a homogeneous mixture of a salt of the present invention.
  • the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, e.g., about 0.1 to about 1000 mg of the active ingredient of the present invention.
  • the tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
  • liquid forms in which the salts and compositions of the present invention can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face mask, tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
  • Topical formulations can contain one or more conventional carriers.
  • ointments can contain water and one or more hydrophobic carriers selected from, e.g., liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like.
  • Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g., glycerinemonostearate, PEG-glycerinemonostearate and cetylstearyl alcohol.
  • Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, e.g., glycerol, hydroxyethyl cellulose, and the like.
  • topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2 or at least about 5 wt % of the salt of the invention.
  • the topical formulations can be suitably packaged in tubes of, e.g., 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.
  • compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient and the like.
  • compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8.
  • the therapeutic dosage of a salt of the present invention can vary according to, e.g., the particular use for which the treatment is made, the manner of administration of the salt, the health and condition of the patient, and the judgment of the prescribing physician.
  • the proportion or concentration of a salt of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration.
  • the salts of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the salt for parenteral administration. Some typical dose ranges are from about 1 ⁇ g/kg to about 1 g/kg of body weight per day.
  • the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the salt selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the salts of the present disclosure can further be useful in investigations of biological processes in normal and abnormal tissues.
  • another aspect of the present invention relates to labeled salts of the invention (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating PD-1 or PD-L1 protein in tissue samples, including human, and for identifying PD-L1 ligands by inhibition binding of a labeled compound.
  • the present invention includes PD-1/PD-L1 binding assays that contain such labeled salts.
  • the present invention further includes isotopically-substituted salts of the disclosure.
  • An “isotopically-substituted” salt is a salt of the invention where one or more atoms are replaced or substituted by an atom having the same atomic number but a different atomic mass or mass number, e.g., a different atomic mass or mass number from the atomic mass or mass number typically found in nature (i.e., naturally occurring).
  • a “radio-labeled” salt is a salt that has incorporated at least one isotope that is radioactive (e.g., radionuclide).
  • Suitable radionuclides that may be incorporated in salts of the present invention include but are not limited to 3 H (also written as T for tritium), 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 18 F, 35 S, 36 Cl, 82 Br, 75 Br, 76 Br, 77 Br, 123 I, 124 I, 125 I and 131 I.
  • the radionuclide that is incorporated in the instant radio-labeled salts will depend on the specific application of that radio-labeled salt. For example, for in vitro PD-L1 protein labeling and competition assays, salts that incorporate 3 H, 14 C, 82 Br, 125 , 131 I, 35 S or will generally be most useful. For radio-imaging applications 11 C, 18 F, 125 I, 123 I, 124 I, 131 I, 75 Br, 76 Br or 77 Br will generally be most useful.
  • the radionuclide is selected from the group consisting of 3 H, 14 C, 125 I, 35 S and 82 Br. Synthetic methods for incorporating radio-isotopes into organic compounds and salts are known in the art.
  • a labeled salt of the invention can be used in a screening assay to identify and/or evaluate compounds.
  • a newly synthesized or identified salt i.e., test salt
  • a test salt which is labeled can be evaluated for its ability to bind a PD-L1 protein by monitoring its concentration variation when contacting with the PD-L1 protein, through tracking of the labeling.
  • a test salt (labeled) can be evaluated for its ability to reduce binding of another compound which is known to bind to a PD-L1 protein (i.e., standard compound). Accordingly, the ability of a test salt to compete with the standard compound for binding to the PD-L1 protein directly correlates to its binding affinity.
  • the standard salt is labeled and test compounds are unlabeled. Accordingly, the concentration of the labeled standard compound is monitored in order to evaluate the competition between the standard compound and the test salt, and the relative binding affinity of the test salt is thus ascertained.
  • kits useful useful, e.g., in the treatment or prevention of diseases or disorders associated with the activity of PD-L1 including its interaction with other proteins such as PD-1 and B7-1 (CD80), such as cancer or infections, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a salt of Compound 1, or any of the embodiments thereof.
  • kits can further include one or more of various conventional pharmaceutical kit components, such as, e.g., containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • X-Ray Powder Diffraction analysis was carried out on a Rigaku MiniFlex X-ray Powder Diffractometer (XRPD) instrument.
  • the general experimental procedures for XRPD were: (1) X-ray radiation from copper at 1.054056 ⁇ with K ⁇ filter; (2) X-ray power at 30 KV, 15 mA; and (3) the sample powder was dispersed on a zero-background sample holder.
  • the general measurement conditions for XRPD were: Start Angle 3 degrees; Stop Angle 45 degrees; Sampling 0.02 degrees; and Scan speed 2 degree/min.
  • DSC Differential Scanning Calorimetry
  • TGA Thermogravimetric analysis
  • Thermogravimetric analysis was carried out on a TA Instrument Thermogravimetric Analyzer, Model Q500.
  • the general experimental conditions for TGA were: ramp from 20° C.-600° C. at 20° C./min; nitrogen purge, gas flow at 40 mL/min followed by balance of the purge flow; sample purge flow at 60 mL/min; platinum sample pan.
  • the solution (slightly turbid) was polish filtered directly into a 60 mL addition funnel using a syringe and filter disks.
  • the round bottom flask was rinsed with 6 mL 10% methanol/dichloromethane solution and the solution was filtered into the addition funnel.
  • the Compound 1 solution was added over 29 minutes.
  • the addition funnel was rinsed with 4 mL 10% methanol/dichloromethane, and added to the slurry. The slurry was stirred for 90 minutes at room temperature.
  • the stoichiometric ratio between Compound 1 and hydrobromic acid was determined as 1:1 by HPLC.
  • Compound 1 mono-hydrobromic acid salt was characterized by XRPD ( FIG. 1 ). Analytical data collected on the product were obtained. The purity of the mono-hydrobromic acid salt was determined by HPLC as 99.4%.
  • the stoichiometric ratio between Compound 1 and hydrobromic acid was determined as 1:1 by HPLC.
  • Compound 1 mono-hydrobromic acid salt was characterized by XRPD ( FIG. 2 ). Analytical data collected on the product were obtained. The purity of the mono-hydrobromic acid salt was determined by HPLC as 99.7%.
  • Oxalic acid (7.41 mg, 0.082 mmol, 1.05 eq.) was added to a solution of Compound 1 (54.36 mg, 0.076 mmol, 1.0 eq.) in tetrahydrofuran (2.0 mL).
  • the reaction mixture was stirred to give a slurry.
  • the slurry was stirred for 2 h, and filtered to give a mother liquid and a solid.
  • the solid was dried under vacuum at 40-41° C. overnight to provide Compound 1 mono-oxalic acid salt as an amorphous solid (54.5 mg, 89% yield).
  • the stoichiometric ratio between Compound 1 and oxalic acid was determined as 1:1 by elemental analysis (calculated for C 43 H 41 N 7 O 8 .2H 2 O: C, 62.99; H, 5.53; N, 11.96 and analysis found C, 62.48; H, 5.33; N, 11.44).
  • Compound 1 mono-oxalic acid salt was characterized by XRPD ( FIG. 3 ). TGA of the salt is provided in FIG. 4 . Analytical data collected on the product were obtained. The purity of the mono-oxalic acid salt was determined by HPLC as 99.3%.
  • the Compound 1 solution was polish filtered directly into a clean scintillation vial using a syringe and filter disks.
  • the Compound 1 solution was added dropwise over 3 minutes via pipet.
  • the slurry was stirred for 90 minutes at room temperature, and the solids were filtered.
  • the filter cake was rinsed with acetone (10 mL), and the solids were dried under high vacuum for 18 hours. 565 mg of product was recovered having a purity 98.58%
  • the stoichiometric ratio between Compound 1 and hydrochloric acid was determined as 1:1 by HPLC.
  • Compound 1 mono-hydrochloric acid salt was characterized by XRPD ( FIG. 5 ). Analytical data collected on the product were obtained. The purity of the mono-hydrochloric acid salt was determined by HPLC as 99.0%.
  • the stoichiometric ratio between Compound 1 and hydrobromic acid was determined as 1:2 by HPLC (sample 2).
  • Compound 1 di-hydrobromic acid salt was characterized by XRPD ( FIG. 8 , samples 1 and 2). Analytical data collected on the product were obtained. The purity of the di-hydrobromic acid salt was determined by HPLC as 97.8% (sample 1) and 97.2% (sample 2).
  • the stoichiometric ratio between Compound 1 and hydrobromic acid was determined as 1:2 by HPLC.
  • Compound 1 di-hydrobromic acid salt was characterized by XRPD ( FIG. 9 ). Analytical data collected on the product were obtained. The purity of the di-hydrobromic acid salt was determined by HPLC as 99.7%.
  • Oxalic acid (20.11 mg, 0.223 mmol, 2.05 eq.) was added to a solution of Compound 1 (75.58 mg, 0.109 mmol, 1.0 eq.) in tetrahydrofuran (2.0 mL). The reaction mixture was stirred to give a slurry. The slurry was stirred continuously for 2 h, and filtered to give a mother liquid and a solid. The solid was dried under vacuum at 40-41° C. overnight to provide Compound 1 di-oxalic acid salt (89.5 mg, 94% yield) as a crystalline solid.
  • the stoichiometric ratio between Compound 1 and oxalic acid was determined as 1:2 by elemental analysis (calculated for C 45 H 43 N 7 O 12 ; C, 61.85; H, 4.96; N, 11.22 and analysis found C, 60.91; H, 5.21; N, 10.73).
  • the crystallinity of the di-oxalic acid salt was confirmed by XRPD ( FIG. 10 , Table 1) and further supported by DSC ( FIG. 11 ), indicating the salt with an onset temperature at 222.23° C. and a peak at 235.26° C. TGA of the di-oxalic acid salt is provided in FIG. 12 , and exhibited approximately 0.7% of weight loss up to about 100° C. Analytical data collected on the product were obtained. The purity of the di-oxalic acid salt was determined by HPLC as 96.10%.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
US16/987,557 2019-08-09 2020-08-07 Salts of a PD-1/PD-L1 inhibitor Active 2041-03-15 US11753406B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/987,557 US11753406B2 (en) 2019-08-09 2020-08-07 Salts of a PD-1/PD-L1 inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962884996P 2019-08-09 2019-08-09
US16/987,557 US11753406B2 (en) 2019-08-09 2020-08-07 Salts of a PD-1/PD-L1 inhibitor

Publications (2)

Publication Number Publication Date
US20210040090A1 US20210040090A1 (en) 2021-02-11
US11753406B2 true US11753406B2 (en) 2023-09-12

Family

ID=72179251

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/987,557 Active 2041-03-15 US11753406B2 (en) 2019-08-09 2020-08-07 Salts of a PD-1/PD-L1 inhibitor

Country Status (8)

Country Link
US (1) US11753406B2 (fr)
EP (1) EP4010342A1 (fr)
JP (1) JP2022544189A (fr)
AR (1) AR119624A1 (fr)
CA (1) CA3150434A1 (fr)
MX (1) MX2022001671A (fr)
TW (1) TW202115059A (fr)
WO (1) WO2021030162A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11866435B2 (en) 2015-12-22 2024-01-09 Incyte Corporation Heterocyclic compounds as immunomodulators

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2928164T3 (es) 2015-10-19 2022-11-15 Incyte Corp Compuestos heterocíclicos como inmunomoduladores
CN109641885B (zh) 2015-11-19 2022-09-30 因赛特公司 作为免疫调节剂的杂环化合物
WO2017192961A1 (fr) 2016-05-06 2017-11-09 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
MA45116A (fr) 2016-05-26 2021-06-02 Incyte Corp Composés hétérocycliques comme immunomodulateurs
SG10202012828TA (en) 2016-06-20 2021-01-28 Incyte Corp Heterocyclic compounds as immunomodulators
MA45669A (fr) 2016-07-14 2019-05-22 Incyte Corp Composés hétérocycliques utilisés comme immunomodulateurs
ES2941716T3 (es) 2016-08-29 2023-05-25 Incyte Corp Compuestos heterocíclicos como inmunomoduladores
MA47120A (fr) 2016-12-22 2021-04-28 Incyte Corp Dérivés pyridine utilisés en tant qu'immunomodulateurs
US10308644B2 (en) 2016-12-22 2019-06-04 Incyte Corporation Heterocyclic compounds as immunomodulators
EP3558989B1 (fr) 2016-12-22 2021-04-14 Incyte Corporation Dérivés de triazolo[1,5-a]pyridine en tant qu'immunomodulateurs
AU2019227607C1 (en) 2018-02-27 2023-07-20 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as A2A / A2B inhibitors
WO2019191707A1 (fr) 2018-03-30 2019-10-03 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
JP7328995B2 (ja) 2018-05-11 2023-08-17 インサイト・コーポレイション Pd-l1免疫調節剤としてのテトラヒドロ-イミダゾ[4,5-c]ピリジン誘導体
CA3100731A1 (fr) 2018-05-18 2019-11-21 Incyte Corporation Derives de pyrimidine fusionnes utilises en tant qu'inhibiteurs de a2a/a2b
US11161850B2 (en) 2018-07-05 2021-11-02 Incyte Corporation Fused pyrazine derivatives as A2A / A2B inhibitors
TWI829857B (zh) 2019-01-29 2024-01-21 美商英塞特公司 作為a2a / a2b抑制劑之吡唑并吡啶及三唑并吡啶
AR119624A1 (es) 2019-08-09 2021-12-29 Incyte Corp Sales de un inhibidor de pd-1 / pd-l1
EP4037773A1 (fr) 2019-09-30 2022-08-10 Incyte Corporation Composés pyrido[3,2-d]pyrimidine en tant qu'immunomodulateurs
CN114829366A (zh) 2019-11-11 2022-07-29 因赛特公司 Pd-1/pd-l1抑制剂的盐及结晶形式
US11760756B2 (en) 2020-11-06 2023-09-19 Incyte Corporation Crystalline form of a PD-1/PD-L1 inhibitor
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
MX2023005362A (es) 2020-11-06 2023-06-22 Incyte Corp Proceso para hacer un inhibidor de proteina de muerte programada 1 (pd-1)/ligando de muerte programada 1 (pd-l1) y sales y formas cristalinas del mismo.
WO2022133176A1 (fr) * 2020-12-18 2022-06-23 Incyte Corporation Formulation orale pour un inhibiteur de pd-l1

Citations (342)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR1425700A (fr) 1965-02-22 1966-01-24 Basf Ag Composés formant des complexes métalliques et procédé pour les préparer et les utiliser
US3272781A (en) 1963-08-07 1966-09-13 American Potash & Chem Corp Boroureas of phosphinoborine polymers
US4208328A (en) 1978-04-27 1980-06-17 General Electric Company Alkyl 3,5-dihydroxy-4-(2-benzothiazolyl)benzoates
US4789711A (en) 1986-12-02 1988-12-06 Ciba-Geigy Corporation Multifunctional epoxide resins
EP0361069A1 (fr) 1988-08-23 1990-04-04 BASF Aktiengesellschaft Benzimidazol-2-anilides d'acides carboxyliques, leurs compositions photoprotectrices pour des matériaux organiques et matériaux organiques stabilisés avec ces anilides
US5077164A (en) 1989-06-21 1991-12-31 Minolta Camera Kabushiki Kaisha Photosensitive member containing an azo dye
JPH0611031A (ja) 1992-04-15 1994-01-21 Mitsubishi Motors Corp 車両用自動変速機の変速制御方法
EP0644460A1 (fr) 1993-09-20 1995-03-22 Fuji Photo Film Co., Ltd. Composition pour photoréserve de type positif
WO1998027108A2 (fr) 1996-12-16 1998-06-25 Fujisawa Pharmaceutical Co., Ltd. Nouveaux composes amide
JPH10316853A (ja) 1997-05-15 1998-12-02 Sumitomo Bakelite Co Ltd 半導体多層配線用層間絶縁膜樹脂組成物及び該絶縁膜の製造方法
WO1999018096A1 (fr) 1997-10-02 1999-04-15 Merck & Co., Inc. Inhibiteurs de la prenyl-proteine transferase
WO1999044992A1 (fr) 1998-03-05 1999-09-10 Nissan Chemical Industries, Ltd. Composes d'anilide et herbicide
JP2000128986A (ja) 1998-10-28 2000-05-09 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール前駆体及びポリベンゾオキサゾール
JP2000128987A (ja) 1998-10-28 2000-05-09 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール前駆体及びポリベンゾオキサゾール
CA2355249A1 (fr) 1998-12-18 2000-06-22 Jason M. Hataye Inhibiteurs de proteases
JP2000212281A (ja) 1999-01-27 2000-08-02 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾ―ル前駆体及びポリベンゾオキサゾ―ル樹脂
US6114497A (en) 1998-10-28 2000-09-05 Sumitomo Bakelite Company Limited Polybenzoxazole resin and precursor thereof
WO2001007409A1 (fr) 1999-07-23 2001-02-01 Astrazeneca Uk Limited Derives de carbazole et leur utilisation en tant que ligands du recepteur de neuropeptide y5
JP2001114893A (ja) 1999-10-15 2001-04-24 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール樹脂およびその前駆体
JP2001163975A (ja) 1999-12-03 2001-06-19 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール樹脂及びその前駆体
WO2001047883A1 (fr) 1999-12-27 2001-07-05 Japan Tobacco Inc. Composes a cycles accoles et leur utilisation comme medicaments
US6297351B1 (en) 1998-12-17 2001-10-02 Sumitomo Bakelite Company Limited Polybenzoxazole resin and precursor thereof
WO2001074815A2 (fr) 2000-03-31 2001-10-11 Ortho Mcneil Pharmaceutical, Inc. Imidazopyridines phenyle-substituees
WO2002000196A2 (fr) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Procede de broyage par voie humide
WO2002014321A1 (fr) 2000-08-11 2002-02-21 The Regents Of The University Of California Utilisation d'inhibiteurs de stat-6 comme agents therapeutiques
US6372907B1 (en) 1999-11-03 2002-04-16 Apptera Corporation Water-soluble rhodamine dye peptide conjugates
WO2002048147A2 (fr) 2000-12-15 2002-06-20 Glaxo Group Limited Composes therapeutiques
US20020082266A1 (en) 2000-04-24 2002-06-27 Michel Gallant Method of treatment using phenyl and biaryl derivatives as prostaglandin E inhibitors and compounds useful therefore
WO2002066477A2 (fr) 2001-02-20 2002-08-29 Astrazeneca Ab Composes
WO2002071827A2 (fr) 2001-03-14 2002-09-19 Eli Lilly And Company Modulateurs des recepteurs x de retinoides
WO2002078700A1 (fr) 2001-03-30 2002-10-10 Smithkline Beecham Corporation Pyralopyridines, leur procede de preparation et leur utilisation en tant que composes therapeutiques
WO2002083672A1 (fr) 2001-04-10 2002-10-24 Smithkline Beecham Corporation Composes de pyrazolopyridine antiviraux
WO2002088124A2 (fr) 2001-04-27 2002-11-07 Smithkline Beecham Corporation Composes therapeutiques
US6521618B2 (en) 2000-03-28 2003-02-18 Wyeth 3-cyanoquinolines, 3-cyano-1,6-naphthyridines, and 3-cyano-1,7-naphthyridines as protein kinase inhibitors
WO2003022845A1 (fr) 2001-09-07 2003-03-20 Smithkline Beecham Corporation Pyrazolo-pyridines servant au traitement d'infections herpetiques
WO2003031587A2 (fr) 2001-10-09 2003-04-17 The Regents Of The University Of California Utilisation d'inhibiteurs de stat-6 comme agents therapeutiques
WO2003030901A1 (fr) 2001-10-09 2003-04-17 Pharmacia & Upjohn Company Carbazoles arylsulfonyl tetrahydro et hexahydro substitues utiles comme ligands du recepteur 5-ht-6
US20030134843A1 (en) 2000-02-01 2003-07-17 Wilfried Lubisch Heterocyclic compounds and their use as parp inhibitors
US20030191115A1 (en) 2001-09-21 2003-10-09 Pinto Donald J.P. Lactam-containing compounds and derivatives thereof as factor Xa inhibitors
JP2003287634A (ja) 2002-01-22 2003-10-10 Sumitomo Bakelite Co Ltd プラスチック光導波路用材料及び光導波路
JP3461397B2 (ja) 1995-01-11 2003-10-27 富士写真フイルム株式会社 ポジ型フオトレジスト組成物
WO2004006906A2 (fr) 2002-07-15 2004-01-22 Combinatorx, Incorporated Methodes de traitement de neoplasmes
US20040018986A1 (en) 2002-04-11 2004-01-29 Janos Pitlik Inhibitors of serine proteases, particularly HCV NS3-NS4A protease
JP2004059761A (ja) 2002-07-30 2004-02-26 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール樹脂、その前駆体及びこれらを用いた光導波路材料並びに光導波路
JP2004091369A (ja) 2002-08-30 2004-03-25 Sumitomo Pharmaceut Co Ltd 新規ビフェニル化合物
US20040058938A1 (en) 2000-12-13 2004-03-25 Oliver Cullmann Use of substituted imidazoazines, novel imidazoazines, methods for the production thereof, and agents containing these compounds
US20040063963A1 (en) 2001-04-19 2004-04-01 Ryuzo Ueno Binaphthol derivative and process for producing the same
WO2004033454A1 (fr) 2002-10-03 2004-04-22 Smithkline Beecham Corporation Composes therapeutiques a base de derives de pyrazolopyridine
WO2004035588A1 (fr) 2002-10-15 2004-04-29 Smithkline Beecham Corporation Composes de pyradazine utiles comme inhibiteurs de gsk-3
US20040082635A1 (en) 2001-06-26 2004-04-29 Hiromasa Hashimoto Fused cyclic compounds and medicinal use thereof
US20040186114A1 (en) 2003-03-10 2004-09-23 Boehringer Ingelheim Pharmaceuticals, Inc. Cytokine inhibitors
WO2004085385A2 (fr) 2003-03-20 2004-10-07 Schering Corporation Ligands de recepteurs de cannabinoides
JP2004294556A (ja) 2003-03-25 2004-10-21 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール樹脂、その前駆体及びこれらを用いた光導波路材料並びに光導波路
WO2004089940A1 (fr) 2003-04-11 2004-10-21 Glenmark Pharmaceuticals S.A. Nouveaux composes heterocycliques pour le traitement des affections inflammatoires et allergiques, leur procede de preparation et compositions pharmaceutiques les contenant
US20040214040A1 (en) 2002-12-30 2004-10-28 Soo-Hyoung Lee Biphenyl derivatives and organic electroluminescent device employing the same
WO2005000833A1 (fr) 2003-05-19 2005-01-06 Irm, Llc Composes immunosuppresseurs et compositions
JP2005002330A (ja) 2003-05-19 2005-01-06 Sumitomo Electric Ind Ltd 光学樹脂材料、光学素子、光モジュール、フッ素化ポリマー前駆体及びフッ素化ポリマー
WO2005005429A1 (fr) 2003-06-30 2005-01-20 Cellular Genomics, Inc. Certaines imidazo[1,2-a]pyrazin-8-ylamines substituees heterocycliques et methodes d'inhibition de la tyrosine kinase de bruton utilisant ces composes
EP1505068A1 (fr) 2002-04-23 2005-02-09 Shionogi & Co., Ltd. DERIVE DE PYRAZOLO(1,5-a)PYRIMIDINE ET INHIBITEUR DE LA NAD(P)H OXYDASE CONTENANT LEDIT DERIVE
WO2005014543A1 (fr) 2003-08-06 2005-02-17 Japan Tobacco Inc. Compose a cycle condense utilise comme inhibiteur de la polymerase du vhc
WO2005014599A1 (fr) 2003-06-04 2005-02-17 Cellular Genomics, Inc. Imidazo[1,2-a]pyrazin-8-ylamines et methodes d'inhibition de la tyrosine kinase de bruton par de tels composes
WO2005023761A2 (fr) 2003-09-11 2005-03-17 Kemia, Inc. Inhibiteurs des cytokines
WO2005034869A2 (fr) 2003-10-08 2005-04-21 Irm Llc Composes et compositions convenant comme inhibiteurs de proteine-kinases
WO2005047290A2 (fr) 2003-11-11 2005-05-26 Cellular Genomics Inc. Certaines imidazo[1,2-a]pyrazin-8-ylamines, procede de fabrication et methode d'utilisation
WO2005063710A1 (fr) 2003-12-23 2005-07-14 Basf Aktiengesellschaft Anilides d'acide 3-trifluoromethylpicolinique et leur utilisation comme fongicides
WO2005077948A1 (fr) 2004-02-16 2005-08-25 Daiichi Pharmaceutical Co., Ltd. Composes heterocycliques fongicides
WO2005079802A1 (fr) 2004-02-12 2005-09-01 Merck & Co., Inc. Amides bipyridyles en tant que modulateurs du récepteur-5 métabotropique du glutamate
WO2005080316A2 (fr) 2004-02-20 2005-09-01 Ucl Biomedica Plc. Modulateur
JP2005248082A (ja) 2004-03-05 2005-09-15 Sumitomo Electric Ind Ltd ポリベンゾオキサゾール樹脂前駆体の製造方法およびポリベンゾオキサゾール樹脂の製造方法
WO2005086808A2 (fr) 2004-03-08 2005-09-22 The University Of North Carolina At Chapel Hill Nouvelles c imidazo[1,2-a]pyridines dicationique et nouvelles 5,6,7,8-tetrahydro-imidazo[1,2a]pyridines utilisee s comme agents antiprotozoaires
WO2005086904A2 (fr) 2004-03-08 2005-09-22 Amgen Inc. Modulation therapeutique de l'activite de recepteur de ppar-gamma
JP2005290301A (ja) 2004-04-02 2005-10-20 Sumitomo Electric Ind Ltd ポリベンゾオキサゾール樹脂前駆体の製造方法およびポリベンゾオキサゾール樹脂の製造方法
WO2005097751A2 (fr) 2004-03-31 2005-10-20 Janssen Pharmaceutica, N.V. Composes heterocycliques non-imidazole
US20050245536A1 (en) 2004-05-03 2005-11-03 Boehringer Ingelheim Pharmaceuticals, Inc. Cytokine inhibitors
WO2005103022A1 (fr) 2004-04-20 2005-11-03 Transtech Pharma, Inc. Derives de thiazole et de pyrimidine substitues, modulateurs des recepteurs de la melanocortine
WO2005105798A1 (fr) 2004-04-30 2005-11-10 Grünenthal GmbH Composes d'imidazo[1,2-a]pyridine substitues et medicaments contenant des composes d'imidazo[1,2-a]pyridine substitues
US20050260126A1 (en) 2002-08-30 2005-11-24 Yukitsuka Kudo Diagnostic probes and remedies for diseases with accumulation of prion protein, and stains for prion protein
US20060004010A1 (en) 2002-07-10 2006-01-05 Hiromu Habashita Ccr4 antagonist and medical use thereof
WO2006034317A2 (fr) 2004-09-21 2006-03-30 Glaxo Group Limited Composes chimiques
WO2006034337A2 (fr) 2004-09-23 2006-03-30 Wyeth Derives de carbazole et de cyclopentaindole destines a traiter une infection par le virus de l'hepatite c
EP1644370A2 (fr) 2003-07-11 2006-04-12 Bristol-Myers Squibb Company Derives de tetrahydroquinoleine constituant des modulateurs des recepteurs des cannabinoides
US20060084650A1 (en) 2004-10-15 2006-04-20 Qing Dong Kinase inhibitors
WO2006053121A2 (fr) 2004-11-10 2006-05-18 Cgi Pharmaceuticals, Inc. Certaines imidazo[1,2-a] pyrazin-8-ylamines et les procédés de fabrication et d'utilisation correspondants
WO2006050803A1 (fr) 2004-11-12 2006-05-18 Bayer Cropscience Ag Derives de pyrazole bicycliques et tricycliques substitues, procedes de production de ces derives et utilisation de ces derives comme herbicides et comme regulateurs de la croissance des plantes
US20060183746A1 (en) 2003-06-04 2006-08-17 Currie Kevin S Certain imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of Bruton's tyrosine kinase by such compounds
WO2006094235A1 (fr) 2005-03-03 2006-09-08 Sirtris Pharmaceuticals, Inc. Composés hétérocycliques liquéfiés et leur utilisation comme modulateurs de sirtuine
WO2006099075A2 (fr) 2005-03-10 2006-09-21 Cgi Pharmaceuticals, Inc. Amides substitues, procede pour les produire et procede pour les utiliser
JP2006290883A (ja) 2005-03-17 2006-10-26 Nippon Nohyaku Co Ltd 置換ヘテロ環カルボン酸アニリド誘導体、その中間体及び農園芸用薬剤並びにその使用方法
WO2006125101A2 (fr) 2005-05-20 2006-11-23 Array Biopharma Inc. Composes inhibiteurs raf et procedes d'utilisation de ceux-ci
US20060270686A1 (en) 2003-08-29 2006-11-30 Martha Kelly Anti-cancer agents and uses thereof
WO2007004954A1 (fr) 2005-07-05 2007-01-11 Ge Healthcare Bio-Sciences Ab Derives de [1, 2, 4] triazolo [1, 5-a] pyrimidine utilises comme adsorbants chromatographiques d'adsorption selective d'igg
WO2007034282A2 (fr) 2005-09-19 2007-03-29 Pfizer Products Inc. Composes de biphenylimidazole utilises comme antagonistes du recepteur du c3a
WO2007038314A2 (fr) 2005-09-22 2007-04-05 Bristol-Myers Squibb Company Composes heterocycliques condenses utilises comme modulateurs des kinases
US20070099938A1 (en) 2003-10-24 2007-05-03 Ono Pharmaceutical Co., Ltd. Antistress drug and medical use thereof
WO2007061764A2 (fr) 2005-11-22 2007-05-31 Merck & Co., Inc. Composes tricycliques utilises comme inhibiteurs de kinases
WO2007067711A2 (fr) 2005-12-08 2007-06-14 Amphora Discovery Corporation Certains types d'entites chimiques, compositions et methode de modulation de trpv1
WO2007069565A1 (fr) 2005-12-12 2007-06-21 Ono Pharmaceutical Co., Ltd. Compose heterocyclique bicyclique
WO2007096764A2 (fr) 2006-02-27 2007-08-30 Glenmark Pharmaceuticals S.A. Dérivés d'hétéroaryles bicycliques en tant que modulateurs des récepteurs cannabinoïdes
WO2007113226A1 (fr) 2006-03-31 2007-10-11 Novartis Ag Composés organiques
WO2007146712A2 (fr) 2006-06-09 2007-12-21 Kemia, Inc. Thérapie à base d'inhibiteurs de cytokine
US7320989B2 (en) 2003-02-28 2008-01-22 Encysive Pharmaceuticals, Inc. Pyridine, pyrimidine, quinoline, quinazoline, and naphthalene urotensin-II receptor antagonists
WO2008011560A2 (fr) 2006-07-20 2008-01-24 Mehmet Kahraman Inhibiteurs de la rho kinase à base de benzothiophène
US20080045536A1 (en) 2005-09-22 2008-02-21 Bristol-Myers Squibb Company Fused heterocyclic compounds useful as kinase modulators
WO2008021745A2 (fr) 2006-08-16 2008-02-21 Itherx Pharmaceuticals, Inc. Inhibiteurs de la pénétration du virus de l'hépatite c
WO2008027812A2 (fr) 2006-08-28 2008-03-06 Forest Laboratories Holdings Limited Dérivés d'imidazopyridine et d'imidazopyrimidine
WO2008033854A1 (fr) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Amides substitués, procédé de production et d'utilisation desdits amides
WO2008033857A2 (fr) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Amides substitués, procédé de production et d'utilisation desdits amides
WO2008033858A2 (fr) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Inhibiteurs de kinase et procédés d'utilisation et d'identification des inhibiteurs de kinase
WO2008032171A1 (fr) 2006-09-11 2008-03-20 Matrix Laboratories Ltd. Nouveaux composés hétérocycliques utiles pour traiter les maladies allergiques et inflammatoires, compositions pharmaceutiques les contenant et procédés servant à les préparer
WO2008057254A2 (fr) 2006-10-27 2008-05-15 Wyeth Composés tricycliques servant d'inhibiteurs des métalloprotéases matricielles
WO2008064317A1 (fr) 2006-11-22 2008-05-29 University Of Medicine And Dentistry Of New Jersey Composés actifs de récepteurs opioïdes lipophiles
WO2008062182A1 (fr) 2006-11-21 2008-05-29 F2G Ltd Dérivés de 2-[(2-substitués)-indolizine-3-yl]-2-oxo-acétamide et leur utilisation comme agents antifongiques
WO2008064318A2 (fr) 2006-11-22 2008-05-29 University Of Medicine And Dentistry Of New Jersey Composés actifs de récepteurs opioïdes périphériques
WO2008071944A1 (fr) 2006-12-14 2008-06-19 Boehringer Ingelheim International Gmbh Benzoxasoles utiles dans le traitement de l'inflammation
US20080153834A1 (en) 2006-09-11 2008-06-26 Blomgren Peter A Certain Substituted Amides, Method of Making, and Method of Use Thereof
WO2008079965A1 (fr) 2006-12-22 2008-07-03 Incyte Corporation Hétérocycles substitués servant d'inhibiteurs de janus kinases
EP1942105A1 (fr) 2005-10-25 2008-07-09 Shionogi Co., Ltd. Derive aminodihydrothiazine
WO2008104279A1 (fr) 2007-02-28 2008-09-04 Sanofi-Aventis Imidazo[1,2-a]azines et leur utilisation comme médicaments
WO2008104077A1 (fr) 2007-02-28 2008-09-04 Methylgene Inc. Inhibiteurs à petite molécule de protéines arginine méthyltransférases (prmt)
WO2008104278A1 (fr) 2007-02-28 2008-09-04 Sanofi-Aventis Imidazo[1,2-a] pyridines et leur utilisation en tant que produits pharmaceutiques
JP2008218327A (ja) 2007-03-07 2008-09-18 Hitachi Ltd 電解質、電解質膜、それを用いた膜電極接合体、燃料電池電源及び燃料電池電源システム
WO2008111299A1 (fr) 2007-03-09 2008-09-18 Daiichi Sankyo Company, Limited Nouveau dérivé de diamide
WO2008114002A1 (fr) 2007-03-22 2008-09-25 Astrazeneca Ab Dérivés de quinoléine destinés au traitement de maladies inflammatoires
WO2008118122A2 (fr) 2006-05-08 2008-10-02 Molecular Neuroimaging, Llc Composés et sondes amyloïdes de ceux-ci pour des utilisations thérapeutiques et en imagerie
WO2008133274A1 (fr) 2007-04-24 2008-11-06 Shionogi & Co., Ltd. Dérivés d'aminodihydrothiazine substitués par des groupes cycliques
WO2008134553A1 (fr) 2007-04-26 2008-11-06 Xenon Pharmaceuticals Inc. Procédés de traitement de maladies associées aux canaux sodiques au moyen de composés bicycliques
US20080280891A1 (en) 2006-06-27 2008-11-13 Locus Pharmaceuticals, Inc. Anti-cancer agents and uses thereof
WO2008141249A1 (fr) 2007-05-10 2008-11-20 Acadia Pharmaceuticals Inc. Imidazol (1,2-a)pyridines et composés associés à activité au niveau des récepteurs cannabinoïdes cb2
US7491245B2 (en) 2004-04-06 2009-02-17 The Procter & Gamble Company Keratin dyeing compounds, keratin dyeing compositions containing them, and use thereof
WO2009027733A1 (fr) 2007-08-24 2009-03-05 Astrazeneca Ab Dérivés de (2-pyridin-3-ylimidazo[1,2-b]pyridazin-6-yl)urée en tant qu'agents antibactériens
WO2009038759A2 (fr) 2007-09-20 2009-03-26 Amgen Inc. Composés de modulation des récepteurs s1p et leur utilisation
WO2009039397A2 (fr) 2007-09-20 2009-03-26 Cgi Pharmaceuticals, Inc. Amides substitués, procédé pour les préparer et procédé pour les utiliser
WO2009059162A1 (fr) 2007-11-02 2009-05-07 Hutchison Medipharma Enterprises Limited Inhibiteurs de la cytokine
WO2009062059A2 (fr) 2007-11-08 2009-05-14 Pharmacopeia, Inc. Purinones et 1h-imidazopyridinones isomères comme inhibiteurs de pkc-thêta
WO2009075830A1 (fr) 2007-12-13 2009-06-18 Merck & Co., Inc. Inhibiteurs de janus kinases
US20090163545A1 (en) 2007-12-21 2009-06-25 University Of Rochester Method For Altering The Lifespan Of Eukaryotic Organisms
WO2009077197A1 (fr) 2007-12-19 2009-06-25 Syngenta Participations Ag Composés insecticides
WO2009079683A1 (fr) 2007-12-21 2009-07-02 The University Of Sydney Ligands de la protéine de translocation
WO2009096202A1 (fr) 2008-01-31 2009-08-06 Konica Minolta Holdings, Inc. Composé aromatique polycyclique halogéné et son procédé de production
WO2009106539A1 (fr) 2008-02-26 2009-09-03 Novartis Ag Composés hétérocycliques comme inhibiteurs de cxcr2
WO2009106597A1 (fr) 2008-02-29 2009-09-03 Æterna Zentaris Gmbh Utilisation d’antagonistes du lhrh à des doses non castratrices
US20090253735A1 (en) 2006-09-22 2009-10-08 Sanofi-Aventis 2-ARYL-6-PHENYLIMIDAZO[1,2-a]PYRIDINE DERIVATIVES, PREPARATION THEREOF AND THERAPEUTIC USE THEREOF
WO2009123986A1 (fr) 2008-03-31 2009-10-08 Takeda Pharmaceutical Company Limited Inhibiteurs de kinase 1 de régulation de signal d'apoptose
US20090281075A1 (en) 2006-02-17 2009-11-12 Pharmacopeia, Inc. Isomeric purinones and 1h-imidazopyridinones as pkc-theta inhibitors
WO2009139576A2 (fr) 2008-05-14 2009-11-19 한국화학연구원 Dérivés de pyridine substitués avec de nouveaux benzoxazoles ou des sels pharmaceutiquement acceptables de ces composés, procédé de préparation de ces composés et compositions pharmaceutiques contenant ces composés comme principes actifs destinées à la prévention et au traitement d'une maladie à croissance cellulaire anormale
WO2009143156A2 (fr) 2008-05-19 2009-11-26 Sepracor Inc. Composés imidazo[1,2-a]pyridines
WO2009146358A1 (fr) 2008-05-29 2009-12-03 Sirtris Pharmaceuticals, Inc. Imidazopyridine et analogues liés en tant que modulateurs de la sirtuine
US20090304821A1 (en) 2006-03-08 2009-12-10 Takeda Pharmaceutical Company Limited Pharmaceutical Combination
WO2010011837A1 (fr) 2008-07-24 2010-01-28 Bristol-Myers Squibb Company Composés hétérocycliques condensés utiles en tant que modulateurs de kinases
WO2010029950A1 (fr) 2008-09-09 2010-03-18 日産化学工業株式会社 Procédé pour produire un composé époxy optiquement actif et un composé sulfoxyde optiquement actif, ligand et complexe pour utilisation dans le procédé, et procédé pour produire le complexe
US7691870B2 (en) 2003-07-11 2010-04-06 Merck Patent Gmbh Benzimidazole carboxamides as raf kinase inhibitors
WO2010056875A1 (fr) 2008-11-12 2010-05-20 Cgi Pharmaceuticals, Inc. Pyridazinones et leur utilisation comme inhibiteurs de la btk
WO2010064020A1 (fr) 2008-12-04 2010-06-10 Proximagen Ltd. Composés imidazopyridines
US20100155712A1 (en) 2008-12-22 2010-06-24 Fujifilm Corporation Organic electroluminescent device
US20100160303A1 (en) 2008-12-19 2010-06-24 Bristol-Myers Squibb Company Carbazole carboxamide compounds useful as kinase inhibitors
WO2010071885A1 (fr) 2008-12-19 2010-06-24 Cephalon, Inc. Pyrrolotriazines en tant qu'inhibiteurs d'alk et de jak2
WO2010075376A2 (fr) 2008-12-23 2010-07-01 Abbott Laboratories Composés antiviraux
WO2010080474A1 (fr) 2008-12-19 2010-07-15 Bristol-Myers Squibb Company Carbazole et inhibiteurs de la carboline kinase
WO2010104306A2 (fr) 2009-03-07 2010-09-16 주식회사 메디젠텍 Compositions pharmaceutiques pour traiter ou prévenir des maladies causées par la translocation de gsk3 du noyau cellulaire au cytoplasme, contenant des composés inhibant la translocation de gsk3 du noyau cellulaire au cytoplasme
JP2010202530A (ja) 2009-02-27 2010-09-16 Tokyo Institute Of Technology 含ヘテロ芳香族化合物および光学材料
EP2233474A1 (fr) 2008-01-18 2010-09-29 Eisai R&D Management Co., Ltd. Dérivé d'aminodihydrothiazine condensée
US20100249151A1 (en) 2007-10-11 2010-09-30 Merck Patent Gesellschaft Beschrankter Haftung Imidazo 1, 2-a pyrimidine derivatives for the treatment of diseases such as diabetes
WO2010115736A2 (fr) 2009-04-02 2010-10-14 Merck Serono S.A. Inhibiteurs de la dihydroorotate déshydrogénase
US20100267775A1 (en) 2007-10-24 2010-10-21 Kenji Negoro Oxadiazolidinedione compound
US20100267778A1 (en) 2003-08-04 2010-10-21 Shinya Kusuda Diphenyl ether compound, process for producing the same, and use
WO2010119264A1 (fr) 2009-04-16 2010-10-21 Centro Nacional De Investigaciones Oncólogicas (Cnio) Imidazopyrazines pour une utilisation en tant qu'inhibiteurs de kinase
US20100292227A1 (en) 2009-05-15 2010-11-18 Boehringer Ingleheim International Gmbh Inhibitors of human immunodeficiency virus replication
CN101891895A (zh) 2010-07-28 2010-11-24 南京航空航天大学 基于桥联双水杨醛结构的苯并噻唑类金属配位聚合物及其制法及应用
CN101910158A (zh) 2007-10-25 2010-12-08 阿斯利康(瑞典)有限公司 可用于治疗细胞增殖性病症的吡啶衍生物和吡嗪衍生物
US7851489B2 (en) 2006-11-08 2010-12-14 Bristol-Myers Squibb Company Pyridinone compounds
WO2011002635A1 (fr) 2009-06-30 2011-01-06 Siga Technologies, Inc. Traitement et prévention d'infections par le virus de la dengue
WO2011008709A1 (fr) 2009-07-13 2011-01-20 Gilead Sciences, Inc. Inhibiteurs de la kinase régulant les signaux de l'apoptose
WO2011018170A2 (fr) 2009-08-14 2011-02-17 Bayer Cropscience Ag Carboxamides pesticides
US20110053915A1 (en) 2007-12-14 2011-03-03 Alla Chem, Llc HETEROCYCLIC INHIBITORS OF AN Hh-SIGNAL CASCADE, MEDICINAL COMPOSITIONS BASED THEREON AND METHODS FOR TREATING DISEASES CAUSED BY THE ABERRANT ACTIVITY OF AN Hh-SIGNAL SYSTEM
US20110062858A1 (en) 2006-07-28 2011-03-17 Novaled Ag Oxazole Triplet Emitters for OLED Applications
US20110065699A1 (en) 2008-03-21 2011-03-17 Sanofi-Aventis POLYSUBSTITUTED DERIVATIVES OF 2-ARYL-6-PHENYL-IMIDAZO[1,2-a]PYRIDINES, AND PREPARATION AND THERAPEUTIC USE THEREOF
US20110065700A1 (en) 2008-03-21 2011-03-17 Sanofi-Aventis POLYSUBSTITUTED DERIVATIVES OF 6-HETEROARYLIMIDAZO[1,2-a]PYRIDINES, AND PREPARATION AND THERAPEUTIC USE THEREOF
US20110065745A1 (en) 2008-03-21 2011-03-17 Sanofi-Aventis Polysubstituted 2-aryl-6-phenylimidazo[1,2-a]pyridine derivatives, and preparation and therapeutic use thereof
CN101993415A (zh) 2010-09-15 2011-03-30 北京韩美药品有限公司 作为Hedgehog通路抑制剂的化合物以及包含该化合物的药物组合物及其应用
WO2011044181A1 (fr) 2009-10-08 2011-04-14 Schering Corporation Composés de dioxyde d'iminothiadiazine comme inhibiteurs de bace, compositions et leur utilisation
WO2011047319A2 (fr) 2009-10-16 2011-04-21 Rib-X Pharmaceuticals, Inc. Composés anti-microbiens et procédés permettant de fabriquer et d'utiliser ces composés
WO2011047129A1 (fr) 2009-10-15 2011-04-21 Southern Research Institute Traitement de maladies neurodégénératives, cause d'amélioration de la mémoire et analyse pour évaluer des composés associés
WO2011050245A1 (fr) 2009-10-23 2011-04-28 Yangbo Feng Hétéroaryles bicycliques formant inhibiteurs de la kinase
WO2011082400A2 (fr) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulateurs du récepteur immunosuppresseur pd-1 et procédés d'utilisation de ceux-ci
WO2011097607A1 (fr) 2010-02-08 2011-08-11 Southern Research Institute Traitement antiviral et dosage pour cribler des antiviraux
WO2011113606A1 (fr) 2010-03-18 2011-09-22 Institut Pasteur Korea Composés anti-infectieux
WO2011117264A1 (fr) 2010-03-26 2011-09-29 F. Hoffmann-La Roche Ag Dérivés de n-(imidazopyrimidin-7-yl)-hétéroarylamide et leur utilisation comme inhibiteurs de pde10a
WO2011140202A2 (fr) 2010-05-04 2011-11-10 Yale University Modulateurs de mif
US20110294781A1 (en) 2008-12-26 2011-12-01 Ajinomoto Co., Inc. Pyrazolo-pyrimidine compounds
EP2402345A1 (fr) 2010-06-29 2012-01-04 Basf Se Composés pyrazoliques bicycliques condenses
WO2012016133A2 (fr) 2010-07-29 2012-02-02 President And Fellows Of Harvard College Inhibiteurs de la ros1 kinase pour le traitement de glioblastome et d'autres cancers déficients en p53
WO2012033735A1 (fr) 2010-09-08 2012-03-15 Bristol-Myers Squibb Company Inhibiteurs de réplication du virus du syndrome immunodéficitaire acquis
WO2012047856A2 (fr) 2010-10-04 2012-04-12 Institute For Hepatitis And Virus Research Nouveaux inhibiteurs de la sécrétion d'antigènes du virus de l'hépatite b
US8163743B2 (en) 2008-06-05 2012-04-24 GlaxoGroupLimited 4-carboxamide indazole derivatives useful as inhibitors of PI3-kinases
WO2012052730A1 (fr) 2010-10-21 2012-04-26 Centro Nacional De Investigaciones Oncológicas (Cnio) Utilisation d'inhibiteurs des pi3k pour le traitement de l'obésité, de la stéatose et du vieillissement
WO2012052745A1 (fr) 2010-10-21 2012-04-26 Centro Nacional De Investigaciones Oncológicas (Cnio) Combinaisons d'inhibiteurs de pi3k avec un second agent antitumoral
WO2012068406A2 (fr) 2010-11-18 2012-05-24 Ligand Pharmaceuticals Incorporated Utilisation de mimétiques d'un facteur de croissance hématopoïétique
WO2012080376A1 (fr) 2010-12-17 2012-06-21 Syngenta Participations Ag Composés insecticides
WO2012088411A1 (fr) 2010-12-22 2012-06-28 Pamlico Pharmaceutical Inc. Dérivés de 2-arylimidazo[1,2-b]pyridazine, 2-phénylimidazo[1,2-a]pyridine, et 2-phénylimidazo[1,2-a]pyrazine
WO2012100342A1 (fr) 2011-01-27 2012-08-02 Université de Montréal Pyrazolopyridine et dérivés de pyrazolopyrimidine en tant que modulateurs du récepteur de la mélanocortine-4
WO2012125886A1 (fr) 2011-03-17 2012-09-20 Bristol-Myers Squibb Company Inhibiteurs de jak3 de type pyrrolopyridazine et leur utilisation pour traiter les maladies inflammatoires et auto-immunes
WO2012129562A2 (fr) 2011-03-24 2012-09-27 The Scripps Research Institute Composés et procédés pour l'induction de la chondrogenèse
WO2012139425A1 (fr) 2011-04-13 2012-10-18 Schering Corporation Iminothiazines 5-substituées et leur monoxydes et dioxydes comme inhibiteurs de bace, leurs compositions et leur utilisation
US20120295884A1 (en) 2011-01-04 2012-11-22 Novartis Ag Complement pathway modulators and uses thereof
WO2012159565A1 (fr) 2011-05-23 2012-11-29 南京英派药业有限公司 6-(arylcarboxamide)imidazo[1,2-a]pyrimidine et 6-(arylcarboxamide)[1,2,4]triazolo[4,3-a]pyrimidine utilisées comme inhibiteur de la voie hedgehog et leur utilisation
WO2012166951A1 (fr) 2011-05-31 2012-12-06 Receptos, Inc. Nouveaux stabilisants et modulateurs du récepteur de glp-1
WO2012168733A1 (fr) 2011-06-10 2012-12-13 Ucl Business Plc 8-amino-imidazo[1,2-a]pyrazines substituées en tant qu'agents antibactériens
US20120323002A1 (en) 2009-12-24 2012-12-20 Ajinomoto Co., Inc. Imidazopyridazine compounds
WO2012175991A1 (fr) 2011-06-24 2012-12-27 Pharminox Limited Composés pentacycliques fusionnés anti-prolifératifs
US20120328569A1 (en) 2010-03-02 2012-12-27 Mccomas Casey Cameron Inhibitors of hepatitis c virus ns5b polymerase
WO2013008095A1 (fr) 2011-07-08 2013-01-17 Novartis Ag Nouveaux dérivés de pyrrolopyrimidine
WO2013033901A1 (fr) 2011-09-08 2013-03-14 Merck Sharp & Dohme Corp. Dérivés benzofuranes substitués par un hétérocycle et leurs procédés d'utilisation pour le traitement de maladies virales
WO2013040528A1 (fr) 2011-09-16 2013-03-21 Microbiotix, Inc. Composés antimicrobiens
US20130096118A1 (en) 2010-06-16 2013-04-18 Chunjian Liu Carboline carboxamide compounds useful as kinase inhibitors
WO2013059594A1 (fr) 2011-10-20 2013-04-25 Sirtris Pharmaceuticals, Inc. Aza-hétérocycles bicycliques substitués et analogues en tant que modulateurs des sirtuines
WO2013057650A1 (fr) 2011-10-21 2013-04-25 Torrent Pharmaceuticals Limited Nouveaux imidazopiridines substitues comme modulateurs recepteurs gpbar1
JP2013084945A (ja) 2011-09-29 2013-05-09 Toyo Ink Sc Holdings Co Ltd 太陽電池封止材用樹脂組成物
US20130131063A1 (en) 2006-08-21 2013-05-23 Infinity Pharmaceuticals, Inc. Compounds and Methods for Inhibiting the Interaction of BCL Proteins with Binding Partners
US20130203754A1 (en) 2010-06-25 2013-08-08 Institute Of Pharmacology And Toxicology Academy Of Military Medical Sciences P.L.A. China 2-Aryl Imidazo[1,2-a]Pyridine-3-Acetamide Derivatives, Preparation Methods and Uses Thereof
WO2013120040A1 (fr) 2012-02-10 2013-08-15 Children's Medical Center Corporation Inhibition d'une voie ciblée pour améliorer la structure, le fonctionnement et l'activité musculaires dans la dystrophie musculaire
WO2013134113A1 (fr) 2012-03-05 2013-09-12 Bristol-Myers Squibb Company Pyrimidines condensées en tant qu'inhibiteurs de la réplication du virus de l'immunodéficience
WO2013157021A1 (fr) 2012-04-20 2013-10-24 Advinus Therapeutics Limited Composés bicycliques, compositions et applications médicinales de ceux-ci
WO2013163404A1 (fr) 2012-04-27 2013-10-31 The Uab Research Foundation Traitement d'infections virales avec arn viraux traduits par un mécanisme non médié par l'ires
WO2014009296A1 (fr) 2012-07-13 2014-01-16 Ucb Pharma S.A. Dérivés d'imidazopyrazine en tant que modulateurs de l'activité tnf
WO2014009295A1 (fr) 2012-07-13 2014-01-16 Ucb Pharma S.A. Dérivés d'imidazopyridine utilisables en tant que modulateurs de l'activité tnf
WO2014017087A1 (fr) 2012-07-25 2014-01-30 杏林製薬株式会社 Dérivé pyrazolopyridine ou un sel pharmacologiquement acceptable de celui-ci
US20140058097A1 (en) 2007-04-24 2014-02-27 Shionogi & Co., Ltd. Pharmaceutical composition for treating alzheimer's disease
WO2014039595A1 (fr) 2012-09-06 2014-03-13 Bristol-Myers Squibb Company Inhibiteurs de jak3 de type imidazopyridazine et leur utilisation pour le traitement de maladies inflammatoires et auto-immunes
US20140088117A1 (en) 2012-09-26 2014-03-27 Genentech, Inc. Cyclic ether pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
WO2014061693A1 (fr) 2012-10-17 2014-04-24 塩野義製薬株式会社 Nouveau dérivé carbocyclique non aromatique ou hétérocyclique non aromatique
WO2014081878A2 (fr) 2012-11-21 2014-05-30 Stategics, Inc. Composés triazolo-pyrimidines substitués pour la modulation de la prolifération, la différentiation et la survie cellulaires
CN103933036A (zh) 2013-01-23 2014-07-23 中国人民解放军军事医学科学院毒物药物研究所 2-芳基咪唑并[1,2-α]吡啶-3-乙酰胺衍生物在制备防治PTSD的药物中的用途
WO2014113388A1 (fr) 2013-01-15 2014-07-24 Incyte Corporation Composés de thiazolecarboxamide et de pyridinecarboxamide utiles comme inhibiteurs de kinases pim
WO2014114532A1 (fr) 2013-01-22 2014-07-31 F. Hoffmann-La Roche Ag Fluoro- [1,3] oxazines servant d'inhibiteurs de la bace1
WO2014121085A1 (fr) 2013-01-31 2014-08-07 Thomas Jefferson University Protéines de fusion à base de pd-l1 et pd-l2 et leurs utilisations
JP2014520866A (ja) 2011-07-19 2014-08-25 メルク・シャープ・アンド・ドーム・コーポレーション Btk阻害薬
WO2014133046A1 (fr) 2013-02-27 2014-09-04 持田製薬株式会社 Nouveau dérivé de pyrazole
WO2014138484A1 (fr) 2013-03-08 2014-09-12 Amgen Inc. Composés à base de 1,3-oxazin-2-amine fusionnée avec du cyclopropyle perfluoré utilisables en tant qu'inhibiteurs de la bêta-sécrétase et leurs procédés d'utilisation
CN104045552A (zh) 2013-03-13 2014-09-17 上海先声药物研究有限公司 作为神经保护剂的药用化合物
WO2014138791A1 (fr) 2013-03-13 2014-09-18 Australian Nuclear Science And Technology Organisation Organismes non humains transgéniques ayant des gènes tspo non fonctionnels
US20140288094A1 (en) 2011-09-22 2014-09-25 Merck Sharp & Dohme Corp Pyrazolopyridyl compounds as aldosterone synthase inhibitors
WO2014151634A1 (fr) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Inhibiteurs macrocycliques des interactions protéine-protéine pd-1/pd-l1 et cd80(b7-1)/pd-l1
WO2014152536A2 (fr) 2013-03-14 2014-09-25 Celtaxsys, Inc. Inhibiteurs de la leucotriène a4 hydrolase
WO2014159959A1 (fr) 2013-03-14 2014-10-02 Bristol-Myers Squibb Company Inhibiteurs de la réplication du virus de l'immunodéficience humaine
WO2014181287A1 (fr) 2013-05-09 2014-11-13 Piramal Enterprises Limited Composés hétérocyclyliques et leurs utilisations
WO2014186035A1 (fr) 2013-03-14 2014-11-20 Curadev Pharma Private Ltd. Inhibiteurs de la voie de la kynurénine
JP2014532066A (ja) 2011-10-13 2014-12-04 ノバルティス アーゲー オキサジン誘導体および神経障害の処置におけるその使用
CN104211726A (zh) 2014-08-11 2014-12-17 中南民族大学 非茂类三齿双核钛配合物、制备方法及用途
WO2014210255A1 (fr) 2013-06-26 2014-12-31 Abbvie Inc. Carboxamides primaires servant d'inhibiteurs de la btk
WO2015000715A1 (fr) 2013-07-02 2015-01-08 Syngenta Participations Ag Hétérocycles bi- ou tricycliques à action pesticide avec substituants soufrés
US20150011751A1 (en) 2012-03-09 2015-01-08 Carna Biosciences, Inc. Novel triazine derivative
WO2015013635A2 (fr) 2013-07-25 2015-01-29 Dana-Farber Cancer Institute, Inc. Inhibiteurs des facteurs de transcription et leurs utilisations
WO2015018940A1 (fr) 2013-08-09 2015-02-12 Fundació Institut Català D'investigació Quimica Composés de bis-salfène et composites de matière carbonée les comprenant
US20150073024A1 (en) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited 1,2,4-Oxadiazole Derivatives as Immunomodulators
WO2015034820A1 (fr) 2013-09-04 2015-03-12 Bristol-Myers Squibb Company Composés utiles comme immunomodulateurs
WO2015033301A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,3,4-oxadiazole et 1,3,4-thiadiazole servant d'immunomodulateurs
WO2015036927A1 (fr) 2013-09-10 2015-03-19 Aurigene Discovery Technologies Limited Dérivés peptidomimétiques d'immunomodulation
US8993604B2 (en) 2009-06-30 2015-03-31 Siga Technologies, Inc. Treatment and prevention of dengue virus infections
EP2871179A1 (fr) 2012-07-03 2015-05-13 ONO Pharmaceutical Co., Ltd. Composé ayant une activité agoniste sur un récepteur de la somatostatine, et leur utilisation à des fins médicales
WO2015086502A1 (fr) 2013-12-09 2015-06-18 Ucb Biopharma Sprl Dérivés de pyrazolopyridine comme modulateurs de l'activité du tnf
WO2015086498A1 (fr) 2013-12-09 2015-06-18 Ucb Biopharma Sprl Dérivés de tétrahydroimidazopyridine substitués comme modulateurs de l'activité du tnf
WO2015086512A1 (fr) 2013-12-09 2015-06-18 Ucb Biopharma Sprl Dérivés d'imidazole et de pyrazole condensés comme modulateurs de l'activité du tnf
WO2015086499A1 (fr) 2013-12-09 2015-06-18 Ucb Biopharma Sprl Dérivés d'imidazopyrimidine à titre de modulateurs de l'activité du tnf
WO2015095337A2 (fr) 2013-12-18 2015-06-25 The Rockefeller University Dérivés pyrazolo [1,5-a]pyrimidine carboxamide pour le traitement de troubles cognitifs
US20150181880A1 (en) 2012-06-18 2015-07-02 Sumitomo Chemical Company, Limited Fused heterocyclic compound
WO2015101622A1 (fr) 2014-01-03 2015-07-09 Bayer Cropscience Ag Nouveaux pyrazolyl-hétéroarylamides utilisables comme pesticides
WO2015120364A1 (fr) 2014-02-10 2015-08-13 Merck Sharp & Dohme Corp. Anticorps qui se lient à la protéine tau humaine et dosage pour quantifier la protéine tau humaine au moyen des anticorps
US20150239868A1 (en) 2014-02-25 2015-08-27 Achillion Pharmaceuticals, Inc. Alkyne Compounds for Treatment of Complement Mediated Disorders
JP2015155397A (ja) 2013-09-30 2015-08-27 日本ポリプロ株式会社 ビフェノール化合物及びそれを用いるオレフィン重合用触媒並びにオレフィン重合体の製造方法
US20150258505A1 (en) 2012-12-03 2015-09-17 Fujifilm Corporation Gas separation membrane, gas separation module, gas separation apparatus, and gas separation method
WO2015150097A1 (fr) 2014-04-04 2015-10-08 Iomet Pharma Ltd Dérivés indolés destinés à être utilisés dans le domaine de la médecine
US20150291549A1 (en) 2014-04-14 2015-10-15 Bristol-Myers Squibb Company Compounds Useful as Immunomodulators
US20150299227A1 (en) 2010-03-04 2015-10-22 Merck Sharp & Dohme Corp. Inhibitors of catechol o-methyl transferase and their use in the treatment of psychotic disorders
JP2015193612A (ja) 2014-03-26 2015-11-05 三井化学株式会社 遷移金属化合物、オレフィン重合用触媒およびオレフィン系重合体の製造方法
WO2015197028A1 (fr) 2014-06-28 2015-12-30 Sunshine Lake Pharma Co., Ltd. Composés en tant qu'inhibiteurs du virus de l'hépatite c (vhc) et leurs utilisations en médecine
US20160015690A1 (en) 2012-04-20 2016-01-21 Gilead Sciences, Inc. Therapeutic compounds
US20160046648A1 (en) 2013-03-14 2016-02-18 The Trustees Of Columbia University In The City Of New York 4-phenylpiperidines, their preparation and use
WO2016044604A1 (fr) 2014-09-17 2016-03-24 Epizyme, Inc. Inhibiteurs de carm1 et leurs utilisations
WO2016057500A1 (fr) 2014-10-06 2016-04-14 Merck Patent Gmbh Composés hétéroaryle servant d'inhibiteurs de la btk, et leurs utilisations
WO2016094688A1 (fr) 2014-12-10 2016-06-16 Massachusetts Institute Of Technology Dérivés fusionnés de 1,3-azole utiles pour le traitement de maladies prolifératives
TW201625527A (zh) 2014-09-19 2016-07-16 財團法人臺灣基督長老教會馬偕紀念社會事業基金會馬偕紀念醫院 苯并雜環化合物及其應用
US9394365B1 (en) 2014-03-12 2016-07-19 Yeda Research And Development Co., Ltd Reducing systemic regulatory T cell levels or activity for treatment of alzheimer's disease
JP2016135778A (ja) 2015-01-16 2016-07-28 大塚製薬株式会社 シアノトリアゾール化合物の医薬用途
WO2016116525A1 (fr) 2015-01-20 2016-07-28 Cynora Gmbh Molécules organiques, notamment pour une utilisation dans des composants opto-électroniques
WO2016118404A1 (fr) 2015-01-20 2016-07-28 Merck Sharp & Dohme Corp. Dioxydes d'iminothiadiazine portant un substituant lié à une amine, utilisés en tant qu'inhibiteurs de bace, compositions les contenant et leur utilisation
US20160229816A1 (en) 2013-07-17 2016-08-11 Otsuka Pharmaceutical Co., Ltd. Cyanotriazole compounds
KR101653560B1 (ko) 2016-02-02 2016-09-12 한국화학연구원 신규한 화합물 또는 이의 약학적으로 허용가능한 염, 및 이를 유효성분으로 함유하는 인플루엔자 바이러스 감염으로 인한 질환의 예방 또는 치료용 약학적 조성물
WO2016156282A1 (fr) 2015-04-02 2016-10-06 Bayer Cropscience Aktiengesellschaft Nouveaux composés de triazole pour contrôler des champignons nocifs phytopathogènes
US9540322B2 (en) 2008-08-18 2017-01-10 Yale University MIF modulators
WO2017035405A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés amino pour le traitement de troubles immunitaires et inflammatoires
KR101715090B1 (ko) 2013-08-28 2017-03-13 한국화학연구원 신규한 화합물 또는 이의 약학적으로 허용가능한 염, 및 이를 유효성분으로 함유하는 인플루엔자 바이러스 감염으로 인한 질환의 예방 또는 치료용 약학적 조성물
KR101717601B1 (ko) 2015-11-10 2017-03-20 한국화학연구원 신규한 화합물 또는 이의 약학적으로 허용가능한 염, 및 이를 유효성분으로 함유하는 인플루엔자 바이러스 감염으로 인한 질환의 예방 또는 치료용 약학적 조성물
US9603950B1 (en) 2015-10-25 2017-03-28 Institute Of Nuclear Energy Research Compounds of imaging agent with HDAC inhibitor for treatment of Alzheimer syndrome and method of synthesis thereof
US20170107216A1 (en) 2015-10-19 2017-04-20 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017066227A1 (fr) 2015-10-15 2017-04-20 Bristol-Myers Squibb Company Composés utiles en tant qu'immunomodulateurs
WO2017070320A1 (fr) 2015-10-21 2017-04-27 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Inhibiteurs allostériques de phényl indole de l'atpase p97
US20170145025A1 (en) 2015-11-19 2017-05-25 Incyte Corporation Heterocyclic compounds as immunomodulators
US20170174679A1 (en) 2015-12-22 2017-06-22 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017106634A1 (fr) 2015-12-17 2017-06-22 Incyte Corporation Dérivés de n-phényl-pyridine-2-carboxamide et leur utilisation comme modulateurs d'interactions protéine/protéine pd-1/pd-l1
WO2017112617A1 (fr) 2015-12-22 2017-06-29 Merck Sharp & Dohme Corp. Dérivés de 4-amino-2-(1h-pyrazolo[3,4-b]pyridin-3-yl)-6-oxo-6,7-dihydro-5h-pyrrolo[2,3-d]pyrimidine et dérivés respectifs de (1h-indazol-3-yl) en tant que modulateurs cgmp pour le traitement de maladies cardiovasculaires
WO2017108569A1 (fr) 2015-12-22 2017-06-29 Syngenta Participations Ag Dérivés de pyrazole à activité pesticide
WO2017109041A1 (fr) 2015-12-22 2017-06-29 Synthon B.V. Composition pharmaceutique comprenant du lénalidomide amorphe et un antioxydant
US20170304282A1 (en) 2016-04-22 2017-10-26 Incyte Corporation Formulations of an lsd1 inhibitor
US20170320875A1 (en) 2016-05-06 2017-11-09 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017205464A1 (fr) 2016-05-26 2017-11-30 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
US20170362253A1 (en) 2016-06-20 2017-12-21 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017223239A1 (fr) 2016-06-21 2017-12-28 X4 Pharmaceuticals, Inc. Inhibiteurs de cxcr4 et leurs utilisations
WO2018013789A1 (fr) 2016-07-14 2018-01-18 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
WO2018026971A1 (fr) 2016-08-03 2018-02-08 Arising International, Llc Composés symétriques ou semi-symétriques utiles comme immunomodulateurs
US20180057486A1 (en) 2016-08-29 2018-03-01 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018045084A1 (fr) 2016-08-30 2018-03-08 Tetraphase Pharmaceuticals, Inc. Composés de tétracycline et méthodes de traitement
US20180179197A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US20180179179A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US20180179201A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US20180177870A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Immunomodulator compounds and methods of use
WO2018116259A1 (fr) 2016-12-21 2018-06-28 Acerta Pharma B.V. Inhibiteurs de type imidazopyrazine de tyrosine kinase de bruton
WO2018119036A1 (fr) 2016-12-21 2018-06-28 Janssen Biotech, Inc. Dérivés de pyrazole en tant qu'inhibiteurs de malt1
US20180179202A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US20180177784A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
CL2018001531A1 (es) 2015-12-22 2018-07-13 Immatics Biotechnologies Gmbh Péptidos y combinación de péptidos para el uso en la inmunoterapia contra el cáncer de mama y otros tipos de cáncer
WO2018195321A1 (fr) 2017-04-20 2018-10-25 Gilead Sciences, Inc. Inhibiteurs pd-1/pd-l1
WO2019023575A1 (fr) 2017-07-28 2019-01-31 Chemocentryx, Inc. Composés immunomodulateurs
WO2019032547A1 (fr) 2017-08-08 2019-02-14 Chemocentryx, Inc. Immunomodulateurs macrocycliques
CL2018003734A1 (es) 2016-06-20 2019-02-15 Novartis Ag Formas cristalinas de compuesto de triazolopirimidina.
WO2019034172A1 (fr) 2017-08-18 2019-02-21 上海轶诺药业有限公司 Composé ayant une activité inhibitrice de pd-l1, sa méthode de préparation et son utilisation
CL2018003697A1 (es) 2016-06-20 2019-05-10 Elanco Us Inc Interferón porcino pegilado y métodos de uso del mismo.
WO2019191707A1 (fr) 2018-03-30 2019-10-03 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
WO2019192506A1 (fr) 2018-04-03 2019-10-10 Betta Pharmaceuticals Co., Ltd Immunomodulateurs, compositions et procédés associés
WO2019204609A1 (fr) 2018-04-19 2019-10-24 Gilead Sciences, Inc. Inhibiteurs pd-1/pd-l1
CA3099994A1 (fr) 2018-05-11 2019-11-04 Incyte Corporation Derives de tetrahydro-imidazo[4,5-c]pyridine en tant qu'immunomodulateurs de pd-l1
WO2020086556A1 (fr) 2018-10-24 2020-04-30 Gilead Sciences, Inc. Inhibiteurs de pd-1/pd-l1
WO2020088357A1 (fr) 2018-11-02 2020-05-07 上海再极医药科技有限公司 Composé de type diphényle, son intermédiaire, son procédé de préparation, sa composition pharmaceutique et ses applications
JP2020514271A (ja) 2016-12-22 2020-05-21 カリセラ バイオサイエンシズ,インコーポレイテッド アルギナーゼ活性を阻害するための組成物及び方法
WO2020156323A1 (fr) 2019-01-31 2020-08-06 Betta Pharmaceuticals Co., Ltd Immunomodulateurs, compositions et procédés associés
US10793505B2 (en) 2013-10-18 2020-10-06 Arkema France Unit and process for purification of crude methyl methacrylate
WO2021030162A1 (fr) 2019-08-09 2021-02-18 Incyte Corporation Sels d'un inhibiteur de pd-1/pd-l1
US20210094976A1 (en) 2019-09-30 2021-04-01 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
US20210139511A1 (en) 2019-11-11 2021-05-13 Incyte Corporation Salts and crystalline forms of a pd-1/pd-l1 inhibitor
US20220144832A1 (en) 2020-11-06 2022-05-12 Incyte Corporation Crystalline form of a pd-1/pd-l1 inhibitor
US20220144831A1 (en) 2020-11-06 2022-05-12 Incyte Corporation Process for making a pd-1/pd-l1 inhibitor and salts and crystalline forms thereof
US20220144830A1 (en) 2020-11-06 2022-05-12 Incyte Corporation Process of preparing a pd-1/pd-l1 inhibitor
US20220193050A1 (en) 2020-12-18 2022-06-23 Incyte Corporation Oral formulation for a pd-l1 inhibitor

Patent Citations (462)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3272781A (en) 1963-08-07 1966-09-13 American Potash & Chem Corp Boroureas of phosphinoborine polymers
FR1425700A (fr) 1965-02-22 1966-01-24 Basf Ag Composés formant des complexes métalliques et procédé pour les préparer et les utiliser
US4208328A (en) 1978-04-27 1980-06-17 General Electric Company Alkyl 3,5-dihydroxy-4-(2-benzothiazolyl)benzoates
US4789711A (en) 1986-12-02 1988-12-06 Ciba-Geigy Corporation Multifunctional epoxide resins
EP0361069A1 (fr) 1988-08-23 1990-04-04 BASF Aktiengesellschaft Benzimidazol-2-anilides d'acides carboxyliques, leurs compositions photoprotectrices pour des matériaux organiques et matériaux organiques stabilisés avec ces anilides
US5077164A (en) 1989-06-21 1991-12-31 Minolta Camera Kabushiki Kaisha Photosensitive member containing an azo dye
JPH0611031A (ja) 1992-04-15 1994-01-21 Mitsubishi Motors Corp 車両用自動変速機の変速制御方法
EP0644460A1 (fr) 1993-09-20 1995-03-22 Fuji Photo Film Co., Ltd. Composition pour photoréserve de type positif
JP3461397B2 (ja) 1995-01-11 2003-10-27 富士写真フイルム株式会社 ポジ型フオトレジスト組成物
WO1998027108A2 (fr) 1996-12-16 1998-06-25 Fujisawa Pharmaceutical Co., Ltd. Nouveaux composes amide
JPH10316853A (ja) 1997-05-15 1998-12-02 Sumitomo Bakelite Co Ltd 半導体多層配線用層間絶縁膜樹脂組成物及び該絶縁膜の製造方法
WO1999018096A1 (fr) 1997-10-02 1999-04-15 Merck & Co., Inc. Inhibiteurs de la prenyl-proteine transferase
WO1999044992A1 (fr) 1998-03-05 1999-09-10 Nissan Chemical Industries, Ltd. Composes d'anilide et herbicide
JP2000128986A (ja) 1998-10-28 2000-05-09 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール前駆体及びポリベンゾオキサゾール
JP2000128987A (ja) 1998-10-28 2000-05-09 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール前駆体及びポリベンゾオキサゾール
US6114497A (en) 1998-10-28 2000-09-05 Sumitomo Bakelite Company Limited Polybenzoxazole resin and precursor thereof
US6297351B1 (en) 1998-12-17 2001-10-02 Sumitomo Bakelite Company Limited Polybenzoxazole resin and precursor thereof
CA2355249A1 (fr) 1998-12-18 2000-06-22 Jason M. Hataye Inhibiteurs de proteases
WO2000035886A2 (fr) 1998-12-18 2000-06-22 Axys Pharmaceuticals, Inc. Inhibiteurs de proteases
CN1344256A (zh) 1998-12-18 2002-04-10 Axys药物公司 (杂)芳基双环杂芳基衍生物,其制备方法和用作蛋白酶抑制剂的用途
US6867200B1 (en) 1998-12-18 2005-03-15 Axys Pharmaceuticals, Inc. (Hetero)aryl-bicyclic heteroaryl derivatives, their preparation and their use as protease inhibitors
JP2000212281A (ja) 1999-01-27 2000-08-02 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾ―ル前駆体及びポリベンゾオキサゾ―ル樹脂
WO2001007409A1 (fr) 1999-07-23 2001-02-01 Astrazeneca Uk Limited Derives de carbazole et leur utilisation en tant que ligands du recepteur de neuropeptide y5
JP2001114893A (ja) 1999-10-15 2001-04-24 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール樹脂およびその前駆体
US6372907B1 (en) 1999-11-03 2002-04-16 Apptera Corporation Water-soluble rhodamine dye peptide conjugates
JP2001163975A (ja) 1999-12-03 2001-06-19 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール樹脂及びその前駆体
WO2001047883A1 (fr) 1999-12-27 2001-07-05 Japan Tobacco Inc. Composes a cycles accoles et leur utilisation comme medicaments
US20030134843A1 (en) 2000-02-01 2003-07-17 Wilfried Lubisch Heterocyclic compounds and their use as parp inhibitors
US6521618B2 (en) 2000-03-28 2003-02-18 Wyeth 3-cyanoquinolines, 3-cyano-1,6-naphthyridines, and 3-cyano-1,7-naphthyridines as protein kinase inhibitors
WO2001074815A2 (fr) 2000-03-31 2001-10-11 Ortho Mcneil Pharmaceutical, Inc. Imidazopyridines phenyle-substituees
US20020082266A1 (en) 2000-04-24 2002-06-27 Michel Gallant Method of treatment using phenyl and biaryl derivatives as prostaglandin E inhibitors and compounds useful therefore
WO2002000196A2 (fr) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Procede de broyage par voie humide
WO2002014321A1 (fr) 2000-08-11 2002-02-21 The Regents Of The University Of California Utilisation d'inhibiteurs de stat-6 comme agents therapeutiques
US20040058938A1 (en) 2000-12-13 2004-03-25 Oliver Cullmann Use of substituted imidazoazines, novel imidazoazines, methods for the production thereof, and agents containing these compounds
WO2002048147A2 (fr) 2000-12-15 2002-06-20 Glaxo Group Limited Composes therapeutiques
WO2002066477A2 (fr) 2001-02-20 2002-08-29 Astrazeneca Ab Composes
WO2002071827A2 (fr) 2001-03-14 2002-09-19 Eli Lilly And Company Modulateurs des recepteurs x de retinoides
WO2002078700A1 (fr) 2001-03-30 2002-10-10 Smithkline Beecham Corporation Pyralopyridines, leur procede de preparation et leur utilisation en tant que composes therapeutiques
WO2002083672A1 (fr) 2001-04-10 2002-10-24 Smithkline Beecham Corporation Composes de pyrazolopyridine antiviraux
US20040063963A1 (en) 2001-04-19 2004-04-01 Ryuzo Ueno Binaphthol derivative and process for producing the same
WO2002088124A2 (fr) 2001-04-27 2002-11-07 Smithkline Beecham Corporation Composes therapeutiques
US20040082635A1 (en) 2001-06-26 2004-04-29 Hiromasa Hashimoto Fused cyclic compounds and medicinal use thereof
WO2003022845A1 (fr) 2001-09-07 2003-03-20 Smithkline Beecham Corporation Pyrazolo-pyridines servant au traitement d'infections herpetiques
US20030191115A1 (en) 2001-09-21 2003-10-09 Pinto Donald J.P. Lactam-containing compounds and derivatives thereof as factor Xa inhibitors
WO2003030901A1 (fr) 2001-10-09 2003-04-17 Pharmacia & Upjohn Company Carbazoles arylsulfonyl tetrahydro et hexahydro substitues utiles comme ligands du recepteur 5-ht-6
WO2003031587A2 (fr) 2001-10-09 2003-04-17 The Regents Of The University Of California Utilisation d'inhibiteurs de stat-6 comme agents therapeutiques
JP2003287634A (ja) 2002-01-22 2003-10-10 Sumitomo Bakelite Co Ltd プラスチック光導波路用材料及び光導波路
US20040018986A1 (en) 2002-04-11 2004-01-29 Janos Pitlik Inhibitors of serine proteases, particularly HCV NS3-NS4A protease
EP1505068A1 (fr) 2002-04-23 2005-02-09 Shionogi & Co., Ltd. DERIVE DE PYRAZOLO(1,5-a)PYRIMIDINE ET INHIBITEUR DE LA NAD(P)H OXYDASE CONTENANT LEDIT DERIVE
US20060089362A1 (en) 2002-04-23 2006-04-27 Shionogi & Co., Ltd Pyrazolo [1,5-a] pyrimidine derivative and nad (p) h oxidase inhibitor containing the same
US20060004010A1 (en) 2002-07-10 2006-01-05 Hiromu Habashita Ccr4 antagonist and medical use thereof
WO2004006906A2 (fr) 2002-07-15 2004-01-22 Combinatorx, Incorporated Methodes de traitement de neoplasmes
JP2004059761A (ja) 2002-07-30 2004-02-26 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール樹脂、その前駆体及びこれらを用いた光導波路材料並びに光導波路
US20050260126A1 (en) 2002-08-30 2005-11-24 Yukitsuka Kudo Diagnostic probes and remedies for diseases with accumulation of prion protein, and stains for prion protein
JP2004091369A (ja) 2002-08-30 2004-03-25 Sumitomo Pharmaceut Co Ltd 新規ビフェニル化合物
WO2004033454A1 (fr) 2002-10-03 2004-04-22 Smithkline Beecham Corporation Composes therapeutiques a base de derives de pyrazolopyridine
WO2004035588A1 (fr) 2002-10-15 2004-04-29 Smithkline Beecham Corporation Composes de pyradazine utiles comme inhibiteurs de gsk-3
US20040214040A1 (en) 2002-12-30 2004-10-28 Soo-Hyoung Lee Biphenyl derivatives and organic electroluminescent device employing the same
US7320989B2 (en) 2003-02-28 2008-01-22 Encysive Pharmaceuticals, Inc. Pyridine, pyrimidine, quinoline, quinazoline, and naphthalene urotensin-II receptor antagonists
US20040186114A1 (en) 2003-03-10 2004-09-23 Boehringer Ingelheim Pharmaceuticals, Inc. Cytokine inhibitors
WO2004085385A2 (fr) 2003-03-20 2004-10-07 Schering Corporation Ligands de recepteurs de cannabinoides
JP2004294556A (ja) 2003-03-25 2004-10-21 Sumitomo Bakelite Co Ltd ポリベンゾオキサゾール樹脂、その前駆体及びこれらを用いた光導波路材料並びに光導波路
WO2004089940A1 (fr) 2003-04-11 2004-10-21 Glenmark Pharmaceuticals S.A. Nouveaux composes heterocycliques pour le traitement des affections inflammatoires et allergiques, leur procede de preparation et compositions pharmaceutiques les contenant
JP2005002330A (ja) 2003-05-19 2005-01-06 Sumitomo Electric Ind Ltd 光学樹脂材料、光学素子、光モジュール、フッ素化ポリマー前駆体及びフッ素化ポリマー
WO2005000833A1 (fr) 2003-05-19 2005-01-06 Irm, Llc Composes immunosuppresseurs et compositions
US7417065B2 (en) 2003-05-19 2008-08-26 Irm Llc Immunosuppressant compounds and compositions
US20060183746A1 (en) 2003-06-04 2006-08-17 Currie Kevin S Certain imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of Bruton's tyrosine kinase by such compounds
WO2005014599A1 (fr) 2003-06-04 2005-02-17 Cellular Genomics, Inc. Imidazo[1,2-a]pyrazin-8-ylamines et methodes d'inhibition de la tyrosine kinase de bruton par de tels composes
WO2005005429A1 (fr) 2003-06-30 2005-01-20 Cellular Genomics, Inc. Certaines imidazo[1,2-a]pyrazin-8-ylamines substituees heterocycliques et methodes d'inhibition de la tyrosine kinase de bruton utilisant ces composes
US7691870B2 (en) 2003-07-11 2010-04-06 Merck Patent Gmbh Benzimidazole carboxamides as raf kinase inhibitors
EP1644370A2 (fr) 2003-07-11 2006-04-12 Bristol-Myers Squibb Company Derives de tetrahydroquinoleine constituant des modulateurs des recepteurs des cannabinoides
US20100267778A1 (en) 2003-08-04 2010-10-21 Shinya Kusuda Diphenyl ether compound, process for producing the same, and use
WO2005014543A1 (fr) 2003-08-06 2005-02-17 Japan Tobacco Inc. Compose a cycle condense utilise comme inhibiteur de la polymerase du vhc
US20060270686A1 (en) 2003-08-29 2006-11-30 Martha Kelly Anti-cancer agents and uses thereof
WO2005023761A2 (fr) 2003-09-11 2005-03-17 Kemia, Inc. Inhibiteurs des cytokines
WO2005034869A2 (fr) 2003-10-08 2005-04-21 Irm Llc Composes et compositions convenant comme inhibiteurs de proteine-kinases
US20050187230A1 (en) 2003-10-08 2005-08-25 Irm Llc Compounds and compositions as protein kinase inhibitors
US20070099938A1 (en) 2003-10-24 2007-05-03 Ono Pharmaceutical Co., Ltd. Antistress drug and medical use thereof
WO2005047290A2 (fr) 2003-11-11 2005-05-26 Cellular Genomics Inc. Certaines imidazo[1,2-a]pyrazin-8-ylamines, procede de fabrication et methode d'utilisation
US20050288295A1 (en) 2003-11-11 2005-12-29 Currie Kevin S Certain imidazo[1,2-a]pyrazin-8-ylamines, method of making, and method of use thereof
WO2005063710A1 (fr) 2003-12-23 2005-07-14 Basf Aktiengesellschaft Anilides d'acide 3-trifluoromethylpicolinique et leur utilisation comme fongicides
WO2005079802A1 (fr) 2004-02-12 2005-09-01 Merck & Co., Inc. Amides bipyridyles en tant que modulateurs du récepteur-5 métabotropique du glutamate
WO2005077948A1 (fr) 2004-02-16 2005-08-25 Daiichi Pharmaceutical Co., Ltd. Composes heterocycliques fongicides
US20070191395A1 (en) 2004-02-16 2007-08-16 Katsuhiro Kawakami Heterocyclic compounds having antifungal activity
WO2005080316A2 (fr) 2004-02-20 2005-09-01 Ucl Biomedica Plc. Modulateur
JP2005248082A (ja) 2004-03-05 2005-09-15 Sumitomo Electric Ind Ltd ポリベンゾオキサゾール樹脂前駆体の製造方法およびポリベンゾオキサゾール樹脂の製造方法
WO2005086808A2 (fr) 2004-03-08 2005-09-22 The University Of North Carolina At Chapel Hill Nouvelles c imidazo[1,2-a]pyridines dicationique et nouvelles 5,6,7,8-tetrahydro-imidazo[1,2a]pyridines utilisee s comme agents antiprotozoaires
WO2005086904A2 (fr) 2004-03-08 2005-09-22 Amgen Inc. Modulation therapeutique de l'activite de recepteur de ppar-gamma
WO2005097751A2 (fr) 2004-03-31 2005-10-20 Janssen Pharmaceutica, N.V. Composes heterocycliques non-imidazole
JP2005290301A (ja) 2004-04-02 2005-10-20 Sumitomo Electric Ind Ltd ポリベンゾオキサゾール樹脂前駆体の製造方法およびポリベンゾオキサゾール樹脂の製造方法
US7491245B2 (en) 2004-04-06 2009-02-17 The Procter & Gamble Company Keratin dyeing compounds, keratin dyeing compositions containing them, and use thereof
WO2005103022A1 (fr) 2004-04-20 2005-11-03 Transtech Pharma, Inc. Derives de thiazole et de pyrimidine substitues, modulateurs des recepteurs de la melanocortine
WO2005105798A1 (fr) 2004-04-30 2005-11-10 Grünenthal GmbH Composes d'imidazo[1,2-a]pyridine substitues et medicaments contenant des composes d'imidazo[1,2-a]pyridine substitues
US20050245536A1 (en) 2004-05-03 2005-11-03 Boehringer Ingelheim Pharmaceuticals, Inc. Cytokine inhibitors
WO2006034317A2 (fr) 2004-09-21 2006-03-30 Glaxo Group Limited Composes chimiques
WO2006034337A2 (fr) 2004-09-23 2006-03-30 Wyeth Derives de carbazole et de cyclopentaindole destines a traiter une infection par le virus de l'hepatite c
US20060084650A1 (en) 2004-10-15 2006-04-20 Qing Dong Kinase inhibitors
WO2006053121A2 (fr) 2004-11-10 2006-05-18 Cgi Pharmaceuticals, Inc. Certaines imidazo[1,2-a] pyrazin-8-ylamines et les procédés de fabrication et d'utilisation correspondants
US20060178367A1 (en) 2004-11-10 2006-08-10 Currie Kevin S Certain imidazo[1,2-a]pyrazin-8-ylamines, method of making, and method of use thereof
WO2006050803A1 (fr) 2004-11-12 2006-05-18 Bayer Cropscience Ag Derives de pyrazole bicycliques et tricycliques substitues, procedes de production de ces derives et utilisation de ces derives comme herbicides et comme regulateurs de la croissance des plantes
WO2006094235A1 (fr) 2005-03-03 2006-09-08 Sirtris Pharmaceuticals, Inc. Composés hétérocycliques liquéfiés et leur utilisation comme modulateurs de sirtuine
US20060229337A1 (en) 2005-03-10 2006-10-12 Brittelli David R Certain substituted amides, method of making, and method of use thereof
WO2006099075A2 (fr) 2005-03-10 2006-09-21 Cgi Pharmaceuticals, Inc. Amides substitues, procede pour les produire et procede pour les utiliser
JP2006290883A (ja) 2005-03-17 2006-10-26 Nippon Nohyaku Co Ltd 置換ヘテロ環カルボン酸アニリド誘導体、その中間体及び農園芸用薬剤並びにその使用方法
WO2006125101A2 (fr) 2005-05-20 2006-11-23 Array Biopharma Inc. Composes inhibiteurs raf et procedes d'utilisation de ceux-ci
WO2007004954A1 (fr) 2005-07-05 2007-01-11 Ge Healthcare Bio-Sciences Ab Derives de [1, 2, 4] triazolo [1, 5-a] pyrimidine utilises comme adsorbants chromatographiques d'adsorption selective d'igg
WO2007034282A2 (fr) 2005-09-19 2007-03-29 Pfizer Products Inc. Composes de biphenylimidazole utilises comme antagonistes du recepteur du c3a
US20080045536A1 (en) 2005-09-22 2008-02-21 Bristol-Myers Squibb Company Fused heterocyclic compounds useful as kinase modulators
WO2007038314A2 (fr) 2005-09-22 2007-04-05 Bristol-Myers Squibb Company Composes heterocycliques condenses utilises comme modulateurs des kinases
US20150210680A1 (en) 2005-10-25 2015-07-30 Shionogi & Co., Ltd. Aminodihydrothiazine derivatives
EP1942105A1 (fr) 2005-10-25 2008-07-09 Shionogi Co., Ltd. Derive aminodihydrothiazine
WO2007061764A2 (fr) 2005-11-22 2007-05-31 Merck & Co., Inc. Composes tricycliques utilises comme inhibiteurs de kinases
WO2007067711A2 (fr) 2005-12-08 2007-06-14 Amphora Discovery Corporation Certains types d'entites chimiques, compositions et methode de modulation de trpv1
US20090281120A1 (en) 2005-12-12 2009-11-12 Ono Pharmaceutical Co., Ltd Bicyclic heterocyclic compound
WO2007069565A1 (fr) 2005-12-12 2007-06-21 Ono Pharmaceutical Co., Ltd. Compose heterocyclique bicyclique
US20090281075A1 (en) 2006-02-17 2009-11-12 Pharmacopeia, Inc. Isomeric purinones and 1h-imidazopyridinones as pkc-theta inhibitors
WO2007096764A2 (fr) 2006-02-27 2007-08-30 Glenmark Pharmaceuticals S.A. Dérivés d'hétéroaryles bicycliques en tant que modulateurs des récepteurs cannabinoïdes
US20090304821A1 (en) 2006-03-08 2009-12-10 Takeda Pharmaceutical Company Limited Pharmaceutical Combination
WO2007113226A1 (fr) 2006-03-31 2007-10-11 Novartis Ag Composés organiques
WO2008118122A2 (fr) 2006-05-08 2008-10-02 Molecular Neuroimaging, Llc Composés et sondes amyloïdes de ceux-ci pour des utilisations thérapeutiques et en imagerie
WO2007146712A2 (fr) 2006-06-09 2007-12-21 Kemia, Inc. Thérapie à base d'inhibiteurs de cytokine
US20080280891A1 (en) 2006-06-27 2008-11-13 Locus Pharmaceuticals, Inc. Anti-cancer agents and uses thereof
WO2008011560A2 (fr) 2006-07-20 2008-01-24 Mehmet Kahraman Inhibiteurs de la rho kinase à base de benzothiophène
US20110062858A1 (en) 2006-07-28 2011-03-17 Novaled Ag Oxazole Triplet Emitters for OLED Applications
WO2008021745A2 (fr) 2006-08-16 2008-02-21 Itherx Pharmaceuticals, Inc. Inhibiteurs de la pénétration du virus de l'hépatite c
US20130131063A1 (en) 2006-08-21 2013-05-23 Infinity Pharmaceuticals, Inc. Compounds and Methods for Inhibiting the Interaction of BCL Proteins with Binding Partners
WO2008027812A2 (fr) 2006-08-28 2008-03-06 Forest Laboratories Holdings Limited Dérivés d'imidazopyridine et d'imidazopyrimidine
US20080153834A1 (en) 2006-09-11 2008-06-26 Blomgren Peter A Certain Substituted Amides, Method of Making, and Method of Use Thereof
US20080139557A1 (en) 2006-09-11 2008-06-12 Blomgren Peter A Certain substituted amides, method of making, and method of use thereof
US20100160292A1 (en) 2006-09-11 2010-06-24 Cgi Pharmaceuticals, Inc Kinase Inhibitors, and Methods of Using and Identifying Kinase Inhibitors
WO2008032171A1 (fr) 2006-09-11 2008-03-20 Matrix Laboratories Ltd. Nouveaux composés hétérocycliques utiles pour traiter les maladies allergiques et inflammatoires, compositions pharmaceutiques les contenant et procédés servant à les préparer
WO2008033858A2 (fr) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Inhibiteurs de kinase et procédés d'utilisation et d'identification des inhibiteurs de kinase
WO2008033857A2 (fr) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Amides substitués, procédé de production et d'utilisation desdits amides
WO2008033854A1 (fr) 2006-09-11 2008-03-20 Cgi Pharmaceuticals, Inc. Amides substitués, procédé de production et d'utilisation desdits amides
US20090253735A1 (en) 2006-09-22 2009-10-08 Sanofi-Aventis 2-ARYL-6-PHENYLIMIDAZO[1,2-a]PYRIDINE DERIVATIVES, PREPARATION THEREOF AND THERAPEUTIC USE THEREOF
WO2008057254A2 (fr) 2006-10-27 2008-05-15 Wyeth Composés tricycliques servant d'inhibiteurs des métalloprotéases matricielles
US7851489B2 (en) 2006-11-08 2010-12-14 Bristol-Myers Squibb Company Pyridinone compounds
WO2008062182A1 (fr) 2006-11-21 2008-05-29 F2G Ltd Dérivés de 2-[(2-substitués)-indolizine-3-yl]-2-oxo-acétamide et leur utilisation comme agents antifongiques
WO2008064318A2 (fr) 2006-11-22 2008-05-29 University Of Medicine And Dentistry Of New Jersey Composés actifs de récepteurs opioïdes périphériques
WO2008064317A1 (fr) 2006-11-22 2008-05-29 University Of Medicine And Dentistry Of New Jersey Composés actifs de récepteurs opioïdes lipophiles
WO2008071944A1 (fr) 2006-12-14 2008-06-19 Boehringer Ingelheim International Gmbh Benzoxasoles utiles dans le traitement de l'inflammation
WO2008079965A1 (fr) 2006-12-22 2008-07-03 Incyte Corporation Hétérocycles substitués servant d'inhibiteurs de janus kinases
WO2008104077A1 (fr) 2007-02-28 2008-09-04 Methylgene Inc. Inhibiteurs à petite molécule de protéines arginine méthyltransférases (prmt)
WO2008104279A1 (fr) 2007-02-28 2008-09-04 Sanofi-Aventis Imidazo[1,2-a]azines et leur utilisation comme médicaments
WO2008104278A1 (fr) 2007-02-28 2008-09-04 Sanofi-Aventis Imidazo[1,2-a] pyridines et leur utilisation en tant que produits pharmaceutiques
JP2008218327A (ja) 2007-03-07 2008-09-18 Hitachi Ltd 電解質、電解質膜、それを用いた膜電極接合体、燃料電池電源及び燃料電池電源システム
WO2008111299A1 (fr) 2007-03-09 2008-09-18 Daiichi Sankyo Company, Limited Nouveau dérivé de diamide
WO2008114002A1 (fr) 2007-03-22 2008-09-25 Astrazeneca Ab Dérivés de quinoléine destinés au traitement de maladies inflammatoires
US20140058097A1 (en) 2007-04-24 2014-02-27 Shionogi & Co., Ltd. Pharmaceutical composition for treating alzheimer's disease
WO2008133274A1 (fr) 2007-04-24 2008-11-06 Shionogi & Co., Ltd. Dérivés d'aminodihydrothiazine substitués par des groupes cycliques
WO2008134553A1 (fr) 2007-04-26 2008-11-06 Xenon Pharmaceuticals Inc. Procédés de traitement de maladies associées aux canaux sodiques au moyen de composés bicycliques
WO2008141249A1 (fr) 2007-05-10 2008-11-20 Acadia Pharmaceuticals Inc. Imidazol (1,2-a)pyridines et composés associés à activité au niveau des récepteurs cannabinoïdes cb2
WO2009027733A1 (fr) 2007-08-24 2009-03-05 Astrazeneca Ab Dérivés de (2-pyridin-3-ylimidazo[1,2-b]pyridazin-6-yl)urée en tant qu'agents antibactériens
JP2010540452A (ja) 2007-09-20 2010-12-24 シージーアイ ファーマシューティカルズ,インコーポレイティド 置換アミド、その製造方法、及びその使用方法
WO2009038759A2 (fr) 2007-09-20 2009-03-26 Amgen Inc. Composés de modulation des récepteurs s1p et leur utilisation
WO2009039397A2 (fr) 2007-09-20 2009-03-26 Cgi Pharmaceuticals, Inc. Amides substitués, procédé pour les préparer et procédé pour les utiliser
US20100249151A1 (en) 2007-10-11 2010-09-30 Merck Patent Gesellschaft Beschrankter Haftung Imidazo 1, 2-a pyrimidine derivatives for the treatment of diseases such as diabetes
US20100267775A1 (en) 2007-10-24 2010-10-21 Kenji Negoro Oxadiazolidinedione compound
CN101910158A (zh) 2007-10-25 2010-12-08 阿斯利康(瑞典)有限公司 可用于治疗细胞增殖性病症的吡啶衍生物和吡嗪衍生物
WO2009059162A1 (fr) 2007-11-02 2009-05-07 Hutchison Medipharma Enterprises Limited Inhibiteurs de la cytokine
WO2009062059A2 (fr) 2007-11-08 2009-05-14 Pharmacopeia, Inc. Purinones et 1h-imidazopyridinones isomères comme inhibiteurs de pkc-thêta
WO2009075830A1 (fr) 2007-12-13 2009-06-18 Merck & Co., Inc. Inhibiteurs de janus kinases
US20110053915A1 (en) 2007-12-14 2011-03-03 Alla Chem, Llc HETEROCYCLIC INHIBITORS OF AN Hh-SIGNAL CASCADE, MEDICINAL COMPOSITIONS BASED THEREON AND METHODS FOR TREATING DISEASES CAUSED BY THE ABERRANT ACTIVITY OF AN Hh-SIGNAL SYSTEM
WO2009077197A1 (fr) 2007-12-19 2009-06-25 Syngenta Participations Ag Composés insecticides
US20100273832A1 (en) 2007-12-19 2010-10-28 Syngenta Crop Protection, Inc. Insecticidal compounds
US20090163545A1 (en) 2007-12-21 2009-06-25 University Of Rochester Method For Altering The Lifespan Of Eukaryotic Organisms
WO2009079683A1 (fr) 2007-12-21 2009-07-02 The University Of Sydney Ligands de la protéine de translocation
EP2233474A1 (fr) 2008-01-18 2010-09-29 Eisai R&D Management Co., Ltd. Dérivé d'aminodihydrothiazine condensée
US20130203741A1 (en) 2008-01-18 2013-08-08 Eisai R&D Management Co., Ltd. Fused aminodihydrothiazine derivatives
WO2009096202A1 (fr) 2008-01-31 2009-08-06 Konica Minolta Holdings, Inc. Composé aromatique polycyclique halogéné et son procédé de production
WO2009106539A1 (fr) 2008-02-26 2009-09-03 Novartis Ag Composés hétérocycliques comme inhibiteurs de cxcr2
WO2009106597A1 (fr) 2008-02-29 2009-09-03 Æterna Zentaris Gmbh Utilisation d’antagonistes du lhrh à des doses non castratrices
US20110065700A1 (en) 2008-03-21 2011-03-17 Sanofi-Aventis POLYSUBSTITUTED DERIVATIVES OF 6-HETEROARYLIMIDAZO[1,2-a]PYRIDINES, AND PREPARATION AND THERAPEUTIC USE THEREOF
US20110065699A1 (en) 2008-03-21 2011-03-17 Sanofi-Aventis POLYSUBSTITUTED DERIVATIVES OF 2-ARYL-6-PHENYL-IMIDAZO[1,2-a]PYRIDINES, AND PREPARATION AND THERAPEUTIC USE THEREOF
US20110065745A1 (en) 2008-03-21 2011-03-17 Sanofi-Aventis Polysubstituted 2-aryl-6-phenylimidazo[1,2-a]pyridine derivatives, and preparation and therapeutic use thereof
WO2009123986A1 (fr) 2008-03-31 2009-10-08 Takeda Pharmaceutical Company Limited Inhibiteurs de kinase 1 de régulation de signal d'apoptose
WO2009139576A2 (fr) 2008-05-14 2009-11-19 한국화학연구원 Dérivés de pyridine substitués avec de nouveaux benzoxazoles ou des sels pharmaceutiquement acceptables de ces composés, procédé de préparation de ces composés et compositions pharmaceutiques contenant ces composés comme principes actifs destinées à la prévention et au traitement d'une maladie à croissance cellulaire anormale
WO2009143156A2 (fr) 2008-05-19 2009-11-26 Sepracor Inc. Composés imidazo[1,2-a]pyridines
WO2009146358A1 (fr) 2008-05-29 2009-12-03 Sirtris Pharmaceuticals, Inc. Imidazopyridine et analogues liés en tant que modulateurs de la sirtuine
US8163743B2 (en) 2008-06-05 2012-04-24 GlaxoGroupLimited 4-carboxamide indazole derivatives useful as inhibitors of PI3-kinases
US20110124640A1 (en) 2008-07-24 2011-05-26 Bristol-Myers Squibb Company Fused heterocyclic compounds useful as kinase modulators
WO2010011837A1 (fr) 2008-07-24 2010-01-28 Bristol-Myers Squibb Company Composés hétérocycliques condensés utiles en tant que modulateurs de kinases
US10202343B2 (en) 2008-08-18 2019-02-12 Yale University MIF modulators
US9540322B2 (en) 2008-08-18 2017-01-10 Yale University MIF modulators
US9643922B2 (en) 2008-08-18 2017-05-09 Yale University MIF modulators
US20190270706A1 (en) 2008-08-18 2019-09-05 Yale University Mif modulators
WO2010029950A1 (fr) 2008-09-09 2010-03-18 日産化学工業株式会社 Procédé pour produire un composé époxy optiquement actif et un composé sulfoxyde optiquement actif, ligand et complexe pour utilisation dans le procédé, et procédé pour produire le complexe
US20110301145A1 (en) 2008-11-12 2011-12-08 Gilead Connecticut INc. Pyridazinones, method of making, and method of use thereof
WO2010056875A1 (fr) 2008-11-12 2010-05-20 Cgi Pharmaceuticals, Inc. Pyridazinones et leur utilisation comme inhibiteurs de la btk
WO2010064020A1 (fr) 2008-12-04 2010-06-10 Proximagen Ltd. Composés imidazopyridines
WO2010071885A1 (fr) 2008-12-19 2010-06-24 Cephalon, Inc. Pyrrolotriazines en tant qu'inhibiteurs d'alk et de jak2
US20100160303A1 (en) 2008-12-19 2010-06-24 Bristol-Myers Squibb Company Carbazole carboxamide compounds useful as kinase inhibitors
WO2010080474A1 (fr) 2008-12-19 2010-07-15 Bristol-Myers Squibb Company Carbazole et inhibiteurs de la carboline kinase
US20120058996A1 (en) 2008-12-19 2012-03-08 Bristol-Myers Squibb Company Carbazole carboxamide compounds useful as kinase inhibitors
US20100155712A1 (en) 2008-12-22 2010-06-24 Fujifilm Corporation Organic electroluminescent device
WO2010075376A2 (fr) 2008-12-23 2010-07-01 Abbott Laboratories Composés antiviraux
US8541424B2 (en) 2008-12-23 2013-09-24 Abbott Laboratories Anti-viral compounds
US9163017B2 (en) 2008-12-23 2015-10-20 Abbvie Inc. Anti-viral compounds
US20110294781A1 (en) 2008-12-26 2011-12-01 Ajinomoto Co., Inc. Pyrazolo-pyrimidine compounds
JP2010202530A (ja) 2009-02-27 2010-09-16 Tokyo Institute Of Technology 含ヘテロ芳香族化合物および光学材料
WO2010104306A2 (fr) 2009-03-07 2010-09-16 주식회사 메디젠텍 Compositions pharmaceutiques pour traiter ou prévenir des maladies causées par la translocation de gsk3 du noyau cellulaire au cytoplasme, contenant des composés inhibant la translocation de gsk3 du noyau cellulaire au cytoplasme
WO2010115736A2 (fr) 2009-04-02 2010-10-14 Merck Serono S.A. Inhibiteurs de la dihydroorotate déshydrogénase
WO2010119264A1 (fr) 2009-04-16 2010-10-21 Centro Nacional De Investigaciones Oncólogicas (Cnio) Imidazopyrazines pour une utilisation en tant qu'inhibiteurs de kinase
WO2010130034A1 (fr) 2009-05-15 2010-11-18 Boehringer Ingelheim International Gmbh Inhibiteurs de la replication du virus de l'immunodeficience humaine
US20130203747A1 (en) 2009-05-15 2013-08-08 Gilead Sciences, Inc. Inhibitors of human immunodeficiency virus replication
US20100292227A1 (en) 2009-05-15 2010-11-18 Boehringer Ingleheim International Gmbh Inhibitors of human immunodeficiency virus replication
WO2011002635A1 (fr) 2009-06-30 2011-01-06 Siga Technologies, Inc. Traitement et prévention d'infections par le virus de la dengue
US8993604B2 (en) 2009-06-30 2015-03-31 Siga Technologies, Inc. Treatment and prevention of dengue virus infections
US20160130251A1 (en) 2009-07-13 2016-05-12 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitors
WO2011008709A1 (fr) 2009-07-13 2011-01-20 Gilead Sciences, Inc. Inhibiteurs de la kinase régulant les signaux de l'apoptose
WO2011018170A2 (fr) 2009-08-14 2011-02-17 Bayer Cropscience Ag Carboxamides pesticides
WO2011044181A1 (fr) 2009-10-08 2011-04-14 Schering Corporation Composés de dioxyde d'iminothiadiazine comme inhibiteurs de bace, compositions et leur utilisation
US20150307465A1 (en) 2009-10-08 2015-10-29 Merck Sharp & Dohme Corp. Iminothiadiazine Dioxide Compounds as BACE Inhibitors, Compositions and Their Use
WO2011047129A1 (fr) 2009-10-15 2011-04-21 Southern Research Institute Traitement de maladies neurodégénératives, cause d'amélioration de la mémoire et analyse pour évaluer des composés associés
WO2011047319A2 (fr) 2009-10-16 2011-04-21 Rib-X Pharmaceuticals, Inc. Composés anti-microbiens et procédés permettant de fabriquer et d'utiliser ces composés
WO2011050245A1 (fr) 2009-10-23 2011-04-28 Yangbo Feng Hétéroaryles bicycliques formant inhibiteurs de la kinase
US20120323002A1 (en) 2009-12-24 2012-12-20 Ajinomoto Co., Inc. Imidazopyridazine compounds
WO2011082400A2 (fr) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulateurs du récepteur immunosuppresseur pd-1 et procédés d'utilisation de ceux-ci
WO2011097607A1 (fr) 2010-02-08 2011-08-11 Southern Research Institute Traitement antiviral et dosage pour cribler des antiviraux
US20120328569A1 (en) 2010-03-02 2012-12-27 Mccomas Casey Cameron Inhibitors of hepatitis c virus ns5b polymerase
US20150299227A1 (en) 2010-03-04 2015-10-22 Merck Sharp & Dohme Corp. Inhibitors of catechol o-methyl transferase and their use in the treatment of psychotic disorders
WO2011113606A1 (fr) 2010-03-18 2011-09-22 Institut Pasteur Korea Composés anti-infectieux
WO2011117264A1 (fr) 2010-03-26 2011-09-29 F. Hoffmann-La Roche Ag Dérivés de n-(imidazopyrimidin-7-yl)-hétéroarylamide et leur utilisation comme inhibiteurs de pde10a
WO2011140202A2 (fr) 2010-05-04 2011-11-10 Yale University Modulateurs de mif
US20130096118A1 (en) 2010-06-16 2013-04-18 Chunjian Liu Carboline carboxamide compounds useful as kinase inhibitors
US20130203754A1 (en) 2010-06-25 2013-08-08 Institute Of Pharmacology And Toxicology Academy Of Military Medical Sciences P.L.A. China 2-Aryl Imidazo[1,2-a]Pyridine-3-Acetamide Derivatives, Preparation Methods and Uses Thereof
EP2402345A1 (fr) 2010-06-29 2012-01-04 Basf Se Composés pyrazoliques bicycliques condenses
CN101891895A (zh) 2010-07-28 2010-11-24 南京航空航天大学 基于桥联双水杨醛结构的苯并噻唑类金属配位聚合物及其制法及应用
WO2012016133A2 (fr) 2010-07-29 2012-02-02 President And Fellows Of Harvard College Inhibiteurs de la ros1 kinase pour le traitement de glioblastome et d'autres cancers déficients en p53
WO2012033735A1 (fr) 2010-09-08 2012-03-15 Bristol-Myers Squibb Company Inhibiteurs de réplication du virus du syndrome immunodéficitaire acquis
CN101993415A (zh) 2010-09-15 2011-03-30 北京韩美药品有限公司 作为Hedgehog通路抑制剂的化合物以及包含该化合物的药物组合物及其应用
WO2012034363A1 (fr) 2010-09-15 2012-03-22 北京韩美药品有限公司 Composés d'inhibiteurs de la voie de hedgehog, compositions pharmaceutiques les contenant et leurs utilisations
WO2012047856A2 (fr) 2010-10-04 2012-04-12 Institute For Hepatitis And Virus Research Nouveaux inhibiteurs de la sécrétion d'antigènes du virus de l'hépatite b
WO2012052730A1 (fr) 2010-10-21 2012-04-26 Centro Nacional De Investigaciones Oncológicas (Cnio) Utilisation d'inhibiteurs des pi3k pour le traitement de l'obésité, de la stéatose et du vieillissement
WO2012052745A1 (fr) 2010-10-21 2012-04-26 Centro Nacional De Investigaciones Oncológicas (Cnio) Combinaisons d'inhibiteurs de pi3k avec un second agent antitumoral
WO2012068406A2 (fr) 2010-11-18 2012-05-24 Ligand Pharmaceuticals Incorporated Utilisation de mimétiques d'un facteur de croissance hématopoïétique
WO2012080376A1 (fr) 2010-12-17 2012-06-21 Syngenta Participations Ag Composés insecticides
US20130253011A1 (en) 2010-12-17 2013-09-26 Syngenta Participations Ag Insecticidal compounds
WO2012088411A1 (fr) 2010-12-22 2012-06-28 Pamlico Pharmaceutical Inc. Dérivés de 2-arylimidazo[1,2-b]pyridazine, 2-phénylimidazo[1,2-a]pyridine, et 2-phénylimidazo[1,2-a]pyrazine
US20120295884A1 (en) 2011-01-04 2012-11-22 Novartis Ag Complement pathway modulators and uses thereof
WO2012100342A1 (fr) 2011-01-27 2012-08-02 Université de Montréal Pyrazolopyridine et dérivés de pyrazolopyrimidine en tant que modulateurs du récepteur de la mélanocortine-4
WO2012125886A1 (fr) 2011-03-17 2012-09-20 Bristol-Myers Squibb Company Inhibiteurs de jak3 de type pyrrolopyridazine et leur utilisation pour traiter les maladies inflammatoires et auto-immunes
WO2012129562A2 (fr) 2011-03-24 2012-09-27 The Scripps Research Institute Composés et procédés pour l'induction de la chondrogenèse
WO2012139425A1 (fr) 2011-04-13 2012-10-18 Schering Corporation Iminothiazines 5-substituées et leur monoxydes et dioxydes comme inhibiteurs de bace, leurs compositions et leur utilisation
US20140128382A1 (en) 2011-04-13 2014-05-08 Wen-Lian Wu 5-substituted iminothiazines and their mono- and dioxides as bace inhibitors, compositions, and their use
WO2012159565A1 (fr) 2011-05-23 2012-11-29 南京英派药业有限公司 6-(arylcarboxamide)imidazo[1,2-a]pyrimidine et 6-(arylcarboxamide)[1,2,4]triazolo[4,3-a]pyrimidine utilisées comme inhibiteur de la voie hedgehog et leur utilisation
WO2012166951A1 (fr) 2011-05-31 2012-12-06 Receptos, Inc. Nouveaux stabilisants et modulateurs du récepteur de glp-1
WO2012168733A1 (fr) 2011-06-10 2012-12-13 Ucl Business Plc 8-amino-imidazo[1,2-a]pyrazines substituées en tant qu'agents antibactériens
WO2012175991A1 (fr) 2011-06-24 2012-12-27 Pharminox Limited Composés pentacycliques fusionnés anti-prolifératifs
WO2013008095A1 (fr) 2011-07-08 2013-01-17 Novartis Ag Nouveaux dérivés de pyrrolopyrimidine
US20140243306A1 (en) 2011-07-08 2014-08-28 Novartis Ag Novel pyrrolo pyrimidine derivatives
JP2014520866A (ja) 2011-07-19 2014-08-25 メルク・シャープ・アンド・ドーム・コーポレーション Btk阻害薬
WO2013033901A1 (fr) 2011-09-08 2013-03-14 Merck Sharp & Dohme Corp. Dérivés benzofuranes substitués par un hétérocycle et leurs procédés d'utilisation pour le traitement de maladies virales
WO2013040528A1 (fr) 2011-09-16 2013-03-21 Microbiotix, Inc. Composés antimicrobiens
US20140288094A1 (en) 2011-09-22 2014-09-25 Merck Sharp & Dohme Corp Pyrazolopyridyl compounds as aldosterone synthase inhibitors
JP2013084945A (ja) 2011-09-29 2013-05-09 Toyo Ink Sc Holdings Co Ltd 太陽電池封止材用樹脂組成物
JP2014532066A (ja) 2011-10-13 2014-12-04 ノバルティス アーゲー オキサジン誘導体および神経障害の処置におけるその使用
WO2013059594A1 (fr) 2011-10-20 2013-04-25 Sirtris Pharmaceuticals, Inc. Aza-hétérocycles bicycliques substitués et analogues en tant que modulateurs des sirtuines
WO2013057650A1 (fr) 2011-10-21 2013-04-25 Torrent Pharmaceuticals Limited Nouveaux imidazopiridines substitues comme modulateurs recepteurs gpbar1
WO2013120040A1 (fr) 2012-02-10 2013-08-15 Children's Medical Center Corporation Inhibition d'une voie ciblée pour améliorer la structure, le fonctionnement et l'activité musculaires dans la dystrophie musculaire
WO2013134113A1 (fr) 2012-03-05 2013-09-12 Bristol-Myers Squibb Company Pyrimidines condensées en tant qu'inhibiteurs de la réplication du virus de l'immunodéficience
EP2824099A1 (fr) 2012-03-09 2015-01-14 Carna Biosciences Inc. Nouveau dérivé de triazine
US20150011751A1 (en) 2012-03-09 2015-01-08 Carna Biosciences, Inc. Novel triazine derivative
US20160015690A1 (en) 2012-04-20 2016-01-21 Gilead Sciences, Inc. Therapeutic compounds
WO2013157021A1 (fr) 2012-04-20 2013-10-24 Advinus Therapeutics Limited Composés bicycliques, compositions et applications médicinales de ceux-ci
WO2013163404A1 (fr) 2012-04-27 2013-10-31 The Uab Research Foundation Traitement d'infections virales avec arn viraux traduits par un mécanisme non médié par l'ires
US20150181880A1 (en) 2012-06-18 2015-07-02 Sumitomo Chemical Company, Limited Fused heterocyclic compound
EP2871179A1 (fr) 2012-07-03 2015-05-13 ONO Pharmaceutical Co., Ltd. Composé ayant une activité agoniste sur un récepteur de la somatostatine, et leur utilisation à des fins médicales
US20150232478A1 (en) 2012-07-03 2015-08-20 Ono Pharmaceutical Co., Ltd. Compound having agonistic activity on somatostatin receptor, and use thereof for medical purposes
WO2014009296A1 (fr) 2012-07-13 2014-01-16 Ucb Pharma S.A. Dérivés d'imidazopyrazine en tant que modulateurs de l'activité tnf
WO2014009295A1 (fr) 2012-07-13 2014-01-16 Ucb Pharma S.A. Dérivés d'imidazopyridine utilisables en tant que modulateurs de l'activité tnf
WO2014017087A1 (fr) 2012-07-25 2014-01-30 杏林製薬株式会社 Dérivé pyrazolopyridine ou un sel pharmacologiquement acceptable de celui-ci
WO2014039595A1 (fr) 2012-09-06 2014-03-13 Bristol-Myers Squibb Company Inhibiteurs de jak3 de type imidazopyridazine et leur utilisation pour le traitement de maladies inflammatoires et auto-immunes
US20140088117A1 (en) 2012-09-26 2014-03-27 Genentech, Inc. Cyclic ether pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
WO2014061693A1 (fr) 2012-10-17 2014-04-24 塩野義製薬株式会社 Nouveau dérivé carbocyclique non aromatique ou hétérocyclique non aromatique
WO2014081878A2 (fr) 2012-11-21 2014-05-30 Stategics, Inc. Composés triazolo-pyrimidines substitués pour la modulation de la prolifération, la différentiation et la survie cellulaires
US20150258505A1 (en) 2012-12-03 2015-09-17 Fujifilm Corporation Gas separation membrane, gas separation module, gas separation apparatus, and gas separation method
WO2014113388A1 (fr) 2013-01-15 2014-07-24 Incyte Corporation Composés de thiazolecarboxamide et de pyridinecarboxamide utiles comme inhibiteurs de kinases pim
US20150376172A1 (en) 2013-01-22 2015-12-31 Thomas Woltering Fluoro-[1,3]oxazines as bace1 inhibitors
WO2014114532A1 (fr) 2013-01-22 2014-07-31 F. Hoffmann-La Roche Ag Fluoro- [1,3] oxazines servant d'inhibiteurs de la bace1
CN103933036A (zh) 2013-01-23 2014-07-23 中国人民解放军军事医学科学院毒物药物研究所 2-芳基咪唑并[1,2-α]吡啶-3-乙酰胺衍生物在制备防治PTSD的药物中的用途
WO2014121085A1 (fr) 2013-01-31 2014-08-07 Thomas Jefferson University Protéines de fusion à base de pd-l1 et pd-l2 et leurs utilisations
WO2014133046A1 (fr) 2013-02-27 2014-09-04 持田製薬株式会社 Nouveau dérivé de pyrazole
US20140378447A1 (en) 2013-02-27 2014-12-25 Mochida Pharmaceutical Co., Ltd. Novel pyrazole derivative
US9611261B2 (en) 2013-03-08 2017-04-04 Amgen Inc. Perfluorinated cyclopropyl fused 1,3-oxazin-2-amine compounds as beta-secretase inhibitors and methods of use
US9085576B2 (en) 2013-03-08 2015-07-21 Amgen Inc. Perfluorinated cyclopropyl fused 1,3-oxazin-2-amine compounds as beta-secretase inhibitors and methods of use
US20160280695A1 (en) 2013-03-08 2016-09-29 Amgen Inc. Perfluorinated cyclopropyl fused 1,3-oxazin-2-amine compounds as beta-secretase inhibitors and methods of use
WO2014138484A1 (fr) 2013-03-08 2014-09-12 Amgen Inc. Composés à base de 1,3-oxazin-2-amine fusionnée avec du cyclopropyle perfluoré utilisables en tant qu'inhibiteurs de la bêta-sécrétase et leurs procédés d'utilisation
US20140275058A1 (en) 2013-03-08 2014-09-18 Amgen Inc. Perfluorinated cyclopropyl fused 1,3-oxazin-2-amine compounds as beta-secretase inhibitors and methods of use
CN105164121A (zh) 2013-03-08 2015-12-16 美国安进公司 作为β分泌酶抑制剂的全氟化环丙基稠合的1,3-噁嗪-2-胺化合物以及其使用方法
US20150252011A1 (en) 2013-03-08 2015-09-10 Amgen Inc. Perfluorinated cyclopropyl fused 1,3-oxazin-2-amine compounds as beta-secretase inhibitors and methods of use
WO2014138791A1 (fr) 2013-03-13 2014-09-18 Australian Nuclear Science And Technology Organisation Organismes non humains transgéniques ayant des gènes tspo non fonctionnels
CN104045552A (zh) 2013-03-13 2014-09-17 上海先声药物研究有限公司 作为神经保护剂的药用化合物
WO2014152536A2 (fr) 2013-03-14 2014-09-25 Celtaxsys, Inc. Inhibiteurs de la leucotriène a4 hydrolase
WO2014159959A1 (fr) 2013-03-14 2014-10-02 Bristol-Myers Squibb Company Inhibiteurs de la réplication du virus de l'immunodéficience humaine
US20160046648A1 (en) 2013-03-14 2016-02-18 The Trustees Of Columbia University In The City Of New York 4-phenylpiperidines, their preparation and use
WO2014186035A1 (fr) 2013-03-14 2014-11-20 Curadev Pharma Private Ltd. Inhibiteurs de la voie de la kynurénine
WO2014151634A1 (fr) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Inhibiteurs macrocycliques des interactions protéine-protéine pd-1/pd-l1 et cd80(b7-1)/pd-l1
WO2014181287A1 (fr) 2013-05-09 2014-11-13 Piramal Enterprises Limited Composés hétérocyclyliques et leurs utilisations
WO2014210255A1 (fr) 2013-06-26 2014-12-31 Abbvie Inc. Carboxamides primaires servant d'inhibiteurs de la btk
US20150005279A1 (en) 2013-06-26 2015-01-01 Abbvie Inc. Primary carboxamides as btk inhibitors
WO2015000715A1 (fr) 2013-07-02 2015-01-08 Syngenta Participations Ag Hétérocycles bi- ou tricycliques à action pesticide avec substituants soufrés
US20160229816A1 (en) 2013-07-17 2016-08-11 Otsuka Pharmaceutical Co., Ltd. Cyanotriazole compounds
WO2015013635A2 (fr) 2013-07-25 2015-01-29 Dana-Farber Cancer Institute, Inc. Inhibiteurs des facteurs de transcription et leurs utilisations
WO2015018940A1 (fr) 2013-08-09 2015-02-12 Fundació Institut Català D'investigació Quimica Composés de bis-salfène et composites de matière carbonée les comprenant
KR101715090B1 (ko) 2013-08-28 2017-03-13 한국화학연구원 신규한 화합물 또는 이의 약학적으로 허용가능한 염, 및 이를 유효성분으로 함유하는 인플루엔자 바이러스 감염으로 인한 질환의 예방 또는 치료용 약학적 조성물
WO2015034820A1 (fr) 2013-09-04 2015-03-12 Bristol-Myers Squibb Company Composés utiles comme immunomodulateurs
CN105705489A (zh) 2013-09-04 2016-06-22 百时美施贵宝公司 用作免疫调节剂的化合物
US20150073024A1 (en) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited 1,2,4-Oxadiazole Derivatives as Immunomodulators
WO2015033299A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,2,4-oxadiazole utilisés comme immunomodulateurs
WO2015033301A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,3,4-oxadiazole et 1,3,4-thiadiazole servant d'immunomodulateurs
US20160194295A1 (en) 2013-09-06 2016-07-07 Aurigene Discovery Technologies Limited 1,3,4-Oxadiazole and 1,3,4-Thiadiazole Derivatives as Immunomodulators
JP2016532710A (ja) 2013-09-06 2016-10-20 オーリジーン ディスカバリー テクノロジーズ リミテッドAurigene Discovery Technologies Limited 免疫調節剤としての1,2,4−オキサジアゾール誘導体
WO2015036927A1 (fr) 2013-09-10 2015-03-19 Aurigene Discovery Technologies Limited Dérivés peptidomimétiques d'immunomodulation
JP2015155397A (ja) 2013-09-30 2015-08-27 日本ポリプロ株式会社 ビフェノール化合物及びそれを用いるオレフィン重合用触媒並びにオレフィン重合体の製造方法
US10793505B2 (en) 2013-10-18 2020-10-06 Arkema France Unit and process for purification of crude methyl methacrylate
WO2015086499A1 (fr) 2013-12-09 2015-06-18 Ucb Biopharma Sprl Dérivés d'imidazopyrimidine à titre de modulateurs de l'activité du tnf
WO2015086512A1 (fr) 2013-12-09 2015-06-18 Ucb Biopharma Sprl Dérivés d'imidazole et de pyrazole condensés comme modulateurs de l'activité du tnf
WO2015086498A1 (fr) 2013-12-09 2015-06-18 Ucb Biopharma Sprl Dérivés de tétrahydroimidazopyridine substitués comme modulateurs de l'activité du tnf
WO2015086502A1 (fr) 2013-12-09 2015-06-18 Ucb Biopharma Sprl Dérivés de pyrazolopyridine comme modulateurs de l'activité du tnf
WO2015095337A2 (fr) 2013-12-18 2015-06-25 The Rockefeller University Dérivés pyrazolo [1,5-a]pyrimidine carboxamide pour le traitement de troubles cognitifs
WO2015101622A1 (fr) 2014-01-03 2015-07-09 Bayer Cropscience Ag Nouveaux pyrazolyl-hétéroarylamides utilisables comme pesticides
WO2015120364A1 (fr) 2014-02-10 2015-08-13 Merck Sharp & Dohme Corp. Anticorps qui se lient à la protéine tau humaine et dosage pour quantifier la protéine tau humaine au moyen des anticorps
US20150239868A1 (en) 2014-02-25 2015-08-27 Achillion Pharmaceuticals, Inc. Alkyne Compounds for Treatment of Complement Mediated Disorders
US9394365B1 (en) 2014-03-12 2016-07-19 Yeda Research And Development Co., Ltd Reducing systemic regulatory T cell levels or activity for treatment of alzheimer's disease
JP2015193612A (ja) 2014-03-26 2015-11-05 三井化学株式会社 遷移金属化合物、オレフィン重合用触媒およびオレフィン系重合体の製造方法
WO2015150097A1 (fr) 2014-04-04 2015-10-08 Iomet Pharma Ltd Dérivés indolés destinés à être utilisés dans le domaine de la médecine
WO2015160641A2 (fr) 2014-04-14 2015-10-22 Bristol-Myers Squibb Company Composés utiles comme immunomodulateurs
US20150291549A1 (en) 2014-04-14 2015-10-15 Bristol-Myers Squibb Company Compounds Useful as Immunomodulators
WO2015197028A1 (fr) 2014-06-28 2015-12-30 Sunshine Lake Pharma Co., Ltd. Composés en tant qu'inhibiteurs du virus de l'hépatite c (vhc) et leurs utilisations en médecine
CN104211726A (zh) 2014-08-11 2014-12-17 中南民族大学 非茂类三齿双核钛配合物、制备方法及用途
WO2016044604A1 (fr) 2014-09-17 2016-03-24 Epizyme, Inc. Inhibiteurs de carm1 et leurs utilisations
TW201625527A (zh) 2014-09-19 2016-07-16 財團法人臺灣基督長老教會馬偕紀念社會事業基金會馬偕紀念醫院 苯并雜環化合物及其應用
WO2016057500A1 (fr) 2014-10-06 2016-04-14 Merck Patent Gmbh Composés hétéroaryle servant d'inhibiteurs de la btk, et leurs utilisations
US10017520B2 (en) 2014-12-10 2018-07-10 Massachusetts Institute Of Technology Myc modulators and uses thereof
WO2016094688A1 (fr) 2014-12-10 2016-06-16 Massachusetts Institute Of Technology Dérivés fusionnés de 1,3-azole utiles pour le traitement de maladies prolifératives
JP2016135778A (ja) 2015-01-16 2016-07-28 大塚製薬株式会社 シアノトリアゾール化合物の医薬用途
WO2016118404A1 (fr) 2015-01-20 2016-07-28 Merck Sharp & Dohme Corp. Dioxydes d'iminothiadiazine portant un substituant lié à une amine, utilisés en tant qu'inhibiteurs de bace, compositions les contenant et leur utilisation
WO2016116525A1 (fr) 2015-01-20 2016-07-28 Cynora Gmbh Molécules organiques, notamment pour une utilisation dans des composants opto-électroniques
WO2016156282A1 (fr) 2015-04-02 2016-10-06 Bayer Cropscience Aktiengesellschaft Nouveaux composés de triazole pour contrôler des champignons nocifs phytopathogènes
WO2017035405A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés amino pour le traitement de troubles immunitaires et inflammatoires
WO2017066227A1 (fr) 2015-10-15 2017-04-20 Bristol-Myers Squibb Company Composés utiles en tant qu'immunomodulateurs
US20170107216A1 (en) 2015-10-19 2017-04-20 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017070089A1 (fr) 2015-10-19 2017-04-27 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
US20190144439A1 (en) 2015-10-19 2019-05-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US20210347771A1 (en) 2015-10-19 2021-11-11 Incyte Corporation Heterocyclic compounds as immunomodulators
US11407749B2 (en) 2015-10-19 2022-08-09 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017070320A1 (fr) 2015-10-21 2017-04-27 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Inhibiteurs allostériques de phényl indole de l'atpase p97
US9603950B1 (en) 2015-10-25 2017-03-28 Institute Of Nuclear Energy Research Compounds of imaging agent with HDAC inhibitor for treatment of Alzheimer syndrome and method of synthesis thereof
KR101717601B1 (ko) 2015-11-10 2017-03-20 한국화학연구원 신규한 화합물 또는 이의 약학적으로 허용가능한 염, 및 이를 유효성분으로 함유하는 인플루엔자 바이러스 감염으로 인한 질환의 예방 또는 치료용 약학적 조성물
US20170145025A1 (en) 2015-11-19 2017-05-25 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017087777A1 (fr) 2015-11-19 2017-05-26 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
US20210221819A1 (en) 2015-11-19 2021-07-22 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017106634A1 (fr) 2015-12-17 2017-06-22 Incyte Corporation Dérivés de n-phényl-pyridine-2-carboxamide et leur utilisation comme modulateurs d'interactions protéine/protéine pd-1/pd-l1
US20180273519A1 (en) 2015-12-17 2018-09-27 Incyte Corporation Heterocyclic compounds as immunomodulators
US20210363137A1 (en) 2015-12-17 2021-11-25 Incyte Corporation Heterocyclic compounds as immunomodulators
US20170174671A1 (en) 2015-12-17 2017-06-22 Incyte Corporation Heterocyclic compounds as immunomodulators
US20170174679A1 (en) 2015-12-22 2017-06-22 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017112730A1 (fr) 2015-12-22 2017-06-29 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
WO2017112617A1 (fr) 2015-12-22 2017-06-29 Merck Sharp & Dohme Corp. Dérivés de 4-amino-2-(1h-pyrazolo[3,4-b]pyridin-3-yl)-6-oxo-6,7-dihydro-5h-pyrrolo[2,3-d]pyrimidine et dérivés respectifs de (1h-indazol-3-yl) en tant que modulateurs cgmp pour le traitement de maladies cardiovasculaires
JP6911031B2 (ja) 2015-12-22 2021-07-28 インサイト・コーポレイションIncyte Corporation 免疫調節剤としての複素環化合物
CL2018001531A1 (es) 2015-12-22 2018-07-13 Immatics Biotechnologies Gmbh Péptidos y combinación de péptidos para el uso en la inmunoterapia contra el cáncer de mama y otros tipos de cáncer
WO2017108569A1 (fr) 2015-12-22 2017-06-29 Syngenta Participations Ag Dérivés de pyrazole à activité pesticide
US20200407357A1 (en) 2015-12-22 2020-12-31 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017109041A1 (fr) 2015-12-22 2017-06-29 Synthon B.V. Composition pharmaceutique comprenant du lénalidomide amorphe et un antioxydant
KR101653560B1 (ko) 2016-02-02 2016-09-12 한국화학연구원 신규한 화합물 또는 이의 약학적으로 허용가능한 염, 및 이를 유효성분으로 함유하는 인플루엔자 바이러스 감염으로 인한 질환의 예방 또는 치료용 약학적 조성물
US20170304282A1 (en) 2016-04-22 2017-10-26 Incyte Corporation Formulations of an lsd1 inhibitor
US20200172541A1 (en) 2016-05-06 2020-06-04 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017192961A1 (fr) 2016-05-06 2017-11-09 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
US20190071439A1 (en) 2016-05-06 2019-03-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US20210017175A1 (en) 2016-05-06 2021-01-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US20170320875A1 (en) 2016-05-06 2017-11-09 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017205464A1 (fr) 2016-05-26 2017-11-30 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
US20170342060A1 (en) 2016-05-26 2017-11-30 Incyte Corporation Heterocyclic compounds as immunomodulators
US20200181126A1 (en) 2016-05-26 2020-06-11 Incyte Corporation Heterocyclic compounds as immunomodulators
US20210017164A1 (en) 2016-05-26 2021-01-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US20190040082A1 (en) 2016-06-20 2019-02-07 Incyte Corporation Heterocyclic compounds as immunomodulators
CL2018003697A1 (es) 2016-06-20 2019-05-10 Elanco Us Inc Interferón porcino pegilado y métodos de uso del mismo.
US20170362253A1 (en) 2016-06-20 2017-12-21 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017222976A1 (fr) 2016-06-20 2017-12-28 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
US20210317139A1 (en) 2016-06-20 2021-10-14 Incyte Corporation Heterocyclic compounds as immunomodulators
JP2019523231A (ja) 2016-06-20 2019-08-22 インサイト・コーポレイションIncyte Corporation 免疫調節剤としての複素環化合物
CL2018003701A1 (es) 2016-06-20 2019-04-05 Incyte Corp Compuestos heterocíclicos como inmunomoduladores.
US20190062345A1 (en) 2016-06-20 2019-02-28 Incyte Corporation Heterocyclic compounds as immunomodulators
CL2018003734A1 (es) 2016-06-20 2019-02-15 Novartis Ag Formas cristalinas de compuesto de triazolopirimidina.
WO2017223239A1 (fr) 2016-06-21 2017-12-28 X4 Pharmaceuticals, Inc. Inhibiteurs de cxcr4 et leurs utilisations
WO2018013789A1 (fr) 2016-07-14 2018-01-18 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
US20210115025A1 (en) 2016-07-14 2021-04-22 Incyte Corporation Heterocyclic compounds as immunomodulators
US20200283423A1 (en) 2016-07-14 2020-09-10 Incyte Corporation Heterocyclic compounds as immunomodulators
US20180016260A1 (en) 2016-07-14 2018-01-18 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018026971A1 (fr) 2016-08-03 2018-02-08 Arising International, Llc Composés symétriques ou semi-symétriques utiles comme immunomodulateurs
JP2019530732A (ja) 2016-08-03 2019-10-24 アライジング・インターナショナル・インコーポレイテッドArising International, Inc. 免疫モジュレータとして有用な対称または半対称化合物
US20200172533A1 (en) 2016-08-29 2020-06-04 Incyte Corporation Heterocyclic compounds as immunomodulators
US20220089588A1 (en) 2016-08-29 2022-03-24 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018044783A1 (fr) 2016-08-29 2018-03-08 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
US20180057486A1 (en) 2016-08-29 2018-03-01 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018045084A1 (fr) 2016-08-30 2018-03-08 Tetraphase Pharmaceuticals, Inc. Composés de tétracycline et méthodes de traitement
WO2018119036A1 (fr) 2016-12-21 2018-06-28 Janssen Biotech, Inc. Dérivés de pyrazole en tant qu'inhibiteurs de malt1
WO2018116259A1 (fr) 2016-12-21 2018-06-28 Acerta Pharma B.V. Inhibiteurs de type imidazopyrazine de tyrosine kinase de bruton
US20180179197A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US20210002276A1 (en) 2016-12-22 2021-01-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US11465981B2 (en) 2016-12-22 2022-10-11 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018119221A1 (fr) 2016-12-22 2018-06-28 Incyte Corporation Dérivés pyridine utilisés en tant qu'immunomodulateurs
CL2019001744A1 (es) 2016-12-22 2019-10-04 Incyte Corp Derivados de benzoxazol como inmunomoduladores.
US20220213090A1 (en) 2016-12-22 2022-07-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US20190202824A1 (en) 2016-12-22 2019-07-04 Incyte Corporation Heterocyclic compounds as immunomodulators
US20220194931A1 (en) 2016-12-22 2022-06-23 Incyte Corporation Heterocyclic compounds as immunomodulators
US11339149B2 (en) 2016-12-22 2022-05-24 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018119236A1 (fr) 2016-12-22 2018-06-28 Incyte Corporation Dérivés de triazolo[1,5-a]pyridine en tant qu'immunomodulateurs
JP2020504739A (ja) 2016-12-22 2020-02-13 インサイト・コーポレイションIncyte Corporation Pd−l1内在化誘導物質としてのテトラヒドロイミダゾ[4,5−c]ピリジン誘導体
JP2020504737A (ja) 2016-12-22 2020-02-13 インサイト・コーポレイションIncyte Corporation 免疫調節剤としての複素環式化合物
WO2018119266A1 (fr) 2016-12-22 2018-06-28 Incyte Corporation Dérivés de benzooxazole en tant qu'mmunomodulateurs
US20180179202A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US20180177784A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
JP2020514271A (ja) 2016-12-22 2020-05-21 カリセラ バイオサイエンシズ,インコーポレイテッド アルギナーゼ活性を阻害するための組成物及び方法
WO2018119263A1 (fr) 2016-12-22 2018-06-28 Incyte Corporation Composés hétérocycliques utilisés en tant qu'inducteurs de l'internalisation de pd-l1
US10308644B2 (en) 2016-12-22 2019-06-04 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018119224A1 (fr) 2016-12-22 2018-06-28 Incyte Corporation Dérivés de tétrahydro imidazo[4,5-c]pyridine en tant qu'inducteurs d'internalisation de pd-l1
US20180179179A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018119286A1 (fr) 2016-12-22 2018-06-28 Incyte Corporation Composés hétéroaromatiques bicycliques utilisés en tant qu'immunomodulateurs
US20180177870A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Immunomodulator compounds and methods of use
US20210107900A1 (en) 2016-12-22 2021-04-15 Incyte Corporation Heterocyclic compounds as immunomodulators
US20190225601A1 (en) 2016-12-22 2019-07-25 Incyte Corporation Heterocyclic compounds as immunomodulators
US10793565B2 (en) 2016-12-22 2020-10-06 Incyte Corporation Heterocyclic compounds as immunomodulators
US20180179201A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US10800768B2 (en) 2016-12-22 2020-10-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US20200325115A1 (en) 2016-12-22 2020-10-15 Incyte Corporation Heterocyclic compounds as immunomodulators
US10806785B2 (en) 2016-12-22 2020-10-20 Incyte Corporation Immunomodulator compounds and methods of use
US20200397893A1 (en) 2016-12-22 2020-12-24 Incyte Corporation Immunomodulator compounds and methods of use
WO2018195321A1 (fr) 2017-04-20 2018-10-25 Gilead Sciences, Inc. Inhibiteurs pd-1/pd-l1
WO2019023575A1 (fr) 2017-07-28 2019-01-31 Chemocentryx, Inc. Composés immunomodulateurs
WO2019032547A1 (fr) 2017-08-08 2019-02-14 Chemocentryx, Inc. Immunomodulateurs macrocycliques
WO2019034172A1 (fr) 2017-08-18 2019-02-21 上海轶诺药业有限公司 Composé ayant une activité inhibitrice de pd-l1, sa méthode de préparation et son utilisation
US20190300524A1 (en) 2018-03-30 2019-10-03 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2019191707A1 (fr) 2018-03-30 2019-10-03 Incyte Corporation Composés hétérocycliques utilisés comme immunomodulateurs
US10669271B2 (en) 2018-03-30 2020-06-02 Incyte Corporation Heterocyclic compounds as immunomodulators
CL2020002511A1 (es) 2018-03-30 2021-01-29 Incyte Corp Compuestos heterocíclicos como inmunomoduladores.
US20200255424A1 (en) 2018-03-30 2020-08-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US20210380584A1 (en) 2018-03-30 2021-12-09 Incyte Corporation Heterocyclic compounds as immunomodulators
US11124511B2 (en) 2018-03-30 2021-09-21 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2019192506A1 (fr) 2018-04-03 2019-10-10 Betta Pharmaceuticals Co., Ltd Immunomodulateurs, compositions et procédés associés
WO2019204609A1 (fr) 2018-04-19 2019-10-24 Gilead Sciences, Inc. Inhibiteurs pd-1/pd-l1
US20200277309A1 (en) 2018-05-11 2020-09-03 Incyte Corporation Heterocyclic compounds as immunomodulators
US20220348594A1 (en) 2018-05-11 2022-11-03 Incyte Corporation Heterocyclic compounds as immunomodulators
US11414433B2 (en) 2018-05-11 2022-08-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US20210115068A1 (en) 2018-05-11 2021-04-22 Incyte Corporation Heterocyclic compounds as immunomodulators
US10618916B2 (en) 2018-05-11 2020-04-14 Incyte Corporation Heterocyclic compounds as immunomodulators
CA3099994A1 (fr) 2018-05-11 2019-11-04 Incyte Corporation Derives de tetrahydro-imidazo[4,5-c]pyridine en tant qu'immunomodulateurs de pd-l1
US20190345170A1 (en) 2018-05-11 2019-11-14 Incyte Corporation Heterocyclic compounds as immunomodulators
US10906920B2 (en) 2018-05-11 2021-02-02 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2020086556A1 (fr) 2018-10-24 2020-04-30 Gilead Sciences, Inc. Inhibiteurs de pd-1/pd-l1
WO2020088357A1 (fr) 2018-11-02 2020-05-07 上海再极医药科技有限公司 Composé de type diphényle, son intermédiaire, son procédé de préparation, sa composition pharmaceutique et ses applications
WO2020156323A1 (fr) 2019-01-31 2020-08-06 Betta Pharmaceuticals Co., Ltd Immunomodulateurs, compositions et procédés associés
WO2021030162A1 (fr) 2019-08-09 2021-02-18 Incyte Corporation Sels d'un inhibiteur de pd-1/pd-l1
US20220340600A1 (en) 2019-09-30 2022-10-27 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
US20210094976A1 (en) 2019-09-30 2021-04-01 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
US11401279B2 (en) 2019-09-30 2022-08-02 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
US20210139511A1 (en) 2019-11-11 2021-05-13 Incyte Corporation Salts and crystalline forms of a pd-1/pd-l1 inhibitor
US20220144830A1 (en) 2020-11-06 2022-05-12 Incyte Corporation Process of preparing a pd-1/pd-l1 inhibitor
US20220144831A1 (en) 2020-11-06 2022-05-12 Incyte Corporation Process for making a pd-1/pd-l1 inhibitor and salts and crystalline forms thereof
US20220144832A1 (en) 2020-11-06 2022-05-12 Incyte Corporation Crystalline form of a pd-1/pd-l1 inhibitor
US20220193050A1 (en) 2020-12-18 2022-06-23 Incyte Corporation Oral formulation for a pd-l1 inhibitor

Non-Patent Citations (284)

* Cited by examiner, † Cited by third party
Title
Abdellaoui et al., "Palladium-catalyzed non-directed C—H bond arylation of difluorobenzenes and dichlorobenzenes bearing benzoxazole or benzothiazole," Catalysis Communications, 2015, 71:13-16.
Ahmed et al., "Enantioselective Polymerization of Epoxides Using Biaryl-Linked Bimetallic Cobalt Catalysts: A Mechanistic Study," J Am Chem Soc., 2013, 135(50):18901-18911.
Alverez et al., "Structure-Activity Study of Bioisosteric Trifluoromethyl and Pentafluorosulfanyl Indole Inhibitors of the AAA ATPase p97," ACS Med Chem., 2015, 6(12):1225-1230.
Amaya et al., "Synthesis of three-dimensionally arranged bis-biphenol ligand on hexaaryl benzene scaffold and its application for cross-pinacol coupling reaction," Tetrahedron Letters, 2011, 52(35):4567-4569.
Anyika et al., "Point-to-Axial Chirality Transfer-A New Probe for "Sensing" the Absolute Configurations of Monoamines," J Am Chem Soc., 2014, 136(2):550-553.
Argentina Office Action in Argentina Application No. 20170103634, dated Jan. 27, 2022, 7 pages.
Arkin et al., "Small-Molecule Inhibitors of Protein-Protein Interactions: Progressing toward the Reality," Chemistry & Biology, Sep. 2014, 21:1102-1114.
Arkin et al., "Small-Molecule Inhibitors of Protein-Protein Interactions: Progressing Towards the Dream," Nature Reviews, Apr. 2004, 3:301-317.
Artz et al., "Host-guest complexation. 28. Hemispherands with four self-organizing units," J Am Chem Soc., 1984, 106(7):2160-2171.
Atzrodt et al., "The Renaissance of H/D Exchange," Angew Chem Int Ed., 2007, 7744-7765.
Australian Notice of Allowance in Australian Application No. 2017382870, dated Mar. 15, 2022, 4 pages.
Australian Office Action in Australian Application No. 2016358100, dated May 8, 2020, 5 pages.
Azuma et al., "B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells," BLOOD, Apr. 1, 2018, 111(7):3635-3643.
Barakat, "Do We Need Small Molecule Inhibitors for the Immune Checkpoints?" J. Pharma. Care Health Sys., 2014, 1(4):10000e119.
Barber et al, "Restoring function in exhausted CD8 T cells during chronic viral infection," Nature, Feb. 2006, 439:682-687.
Bastin et al., "Salt Selection and Optimisation for Pharmaceutical New Chemical Entities," Org Proc Res Dev., dated Jan. 1, 2000, pp. 4(5):427-435.
Bentley et al., "Antenna Biphenols: Development of Extended Wavelength Chiroptical Reporters," J Org Chem., 2016, 81(3):1185-1191.
Berg, "Modulation of Protein-Protein Interactions with Small Organic Molecules," Angew. Chem. Int. Ed., 2003, 42:2462-2481.
Berge et al., "Pharmaceutical Salts," J. Pharm. Sci., Jan. 1977, 66(1):1-19.
Blank et al, "PD-L1/B7H-1 Inhibits the Effector Phase of Tumor Rejection by T Cell Receptor (TCR) Transgenic CD8+ T Cells," Cancer Res., Feb. 2004, 64(3):1140-5.
Blom et al., "Optimizing Preparative LC-MS Configurations and Methods for Parallel Synthesis Purification", J. Combi. Chem., 2003, 5:670-83.
Blom et al., "Preparative LC-MS Purification: Improved Compound Specific Method Optimization", J. Combi. Chem., Nov. 2004, 6:874-883.
Blom, "Two-Pump At Column Dilution Configuration for Preparative LC-MS", K. Blom, J. Combi. Chem., 2002, 4:295-301.
Brazilian Office Action in Brazilian Application No. BR112018012756-6, dated Jan. 5, 2021, 6 pages.
Bross et al., "Radiation damage to 2-(2′-hydroxyphenyl)benzothiazoles," Radiation Physics and Chemistiy, Jul. 1992, 41:379-387.
Buisman et al., "Chiral Cooperativity in Diastereomeric Diphosphite Ligands: Effects on the Rhodium-Catalyzed Enantioselective Hydroformylation of Styrene," Organometallics, 1997, 16(13):2929-2939.
Caira, "Crystalline Polymorphism of Organic Compounds," Topics in Current Chemistry, Jan. 1, 1998, 198:163-208.
Camara et al., "Multiple dermatofibromas: Dermoscopic patterns," Indian journal of dermatology, 2013, 58(3):243.
Carter et al, "PD-1:PD-L inhibitory pathway affects both CD4+ and CD8+ T cells and is overcome by IL-2," Eur. J. Immunol., 2002, 32(3):634-643.
Chang et al., "Blocking of the PD-1/PD-L1 Interaction by a d-Peptide Antagonist for Cancer Immunotherapy," Angew. Chem. Int. Ed., 2015, 127(40):11926-11930.
Chang et al., "Blocking of the PD-1/PD-L1 Interaction by a d-Peptide Antagonist for Cancer Immunotherapy" Angew. Chem. Int. Ed., 2015, 26 pages; Supporting Information for 127(40):11926-11930.
Chen et al., "Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future," J. Clin. Invest, Sep. 2015, 125(9):3384-3391.
Cheng et al., "Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma," Cell Death and Disease, 2018, 9:422.
Cheng et al., "Recent Advances in Small Molecule Based Cancer Immunotherapy," Eur J Med Chem., 2018, 157:582-598.
Cheng et al., "Structure and Interactions of the Human Programmed Cell Death 1 Receptor," J. Bio. Chem., Apr. 2013, 288(17):11771-11785.
Cheng et al., "Synthetic connections to the aromatic directed metalation reaction. Iterative ortho metalation-cross coupling tactics for the construction of polyphenyls," Tetrahedron Letters, 1978, 28(43):5097-5098.
Chilean Office Action in Chilean Application No. 201801685, dated Aug. 20, 2019, 18 pages.
Chilean Office Action in Chilean Application No. 201803701, dated Nov. 22, 2019, 18 page.
Chilean Office Action in Chilean Application No. 201901744, dated Apr. 14, 2020, 19 pages.
Chilean Office Action in Chilean Application No. 2922-2020, dated Dec. 8, 2021, 21 pages.
Chinese Office Action in Chinese Application No. 201680077700.8, dated Jul. 2, 2021, 23 pages.
Chinese Search Report in Chinese Application No. 201780049752.9, dated Dec. 28, 2020, 5 pages.
Clayden et al., "Conformational Preference and Remote (1,10) Stereocontrol in Biphenyl-2,2′-dicarboxamides," Org. Lett, 2001, 3(26):4133-4136.
Colombian Office Action in Colombian Application No. NC2019/0000386, dated Sep. 25, 2020, 18 pages.
Cram et al., "Host-guest complexation. 26. Cavitands composed of fluorobenzene units bonded in their 2,6-positions to form macrocycles," J Am Chem Soc., 1984, 106(3):695-701.
Cram et al., "Host-guest complexation. 29. Expanded hemispherands," J Am Chem Soc., 1984, 106(11):6386-3292.
Cram et al., "Host-guest complexation. 32. Spherands composed of cyclic urea and anisyl units," J Am Chem Soc., 1984, 106(23):7150-7167.
Cram et al., "Spherand hosts containing cyclic urea units," J Am Chem Soc., 1982, 104(24):6828-6830.
Curis, "Overview and Path for Growth," Aurigene Strategic Collaboration, Jan. 21, 2015, 13 slides.
Database accession No. 1478989-52-4 abstract, Nov. 22, 2013, 1 page.
Database Accession No. 1568738-04-4 abstract, Mar. 14, 2014, 1 page.
Database Accession No. 1580823-55-7 abstract, Apr. 6, 2014, 1 page.
Database Accession No. 1581556-71-9 abstract, Apr. 8, 2014, 1 page.
Database Accession No. 1590700-72-3 abstract, Apr. 27, 2014, 1 page.
Database accession No. 2013:447446 abstract, 2013, 1 page.
De Lucca et al., "Small Molecule Reversible Inhibitors of Bruton's Tyrosine Kinase (BTK): Structure-Activity Relationships Leading to the Identification of 7-(2-Hydroxypropan-2-yl)-4-[2-methyl-3-(4-oxo-3,4-dihydroquinazolin-3-yl)phenyl]-9H-carbazole-1-carboxamide (BMS-935177)," Journal of Medicinal Chemistiy, 2016, 59(17):7915-7935.
Dhanunjayarao et al., "Synthesis and Optical Properties of Salicylaldimine-Based Diboron Complexes," Eur J Inorg Chem., 2014, 3:539-545.
Differding, "AUNP-12—A Novel Peptide Therapeutic Targeting PD-1 Immune Checkpoint Pathway for Cancer Immunotherapy—Structure Activity Relationships & Peptide / Peptidomimetic Analogs," Differding Consulting s.p.r.l. (Belgium), Feb. 26, 2014, 12 pages.
Dolan et al., "PD-1 Pathway Inhibitors: Changing the Landscape of Cancer Immunotherapy," Cancer Control, Jul. 2014, 21(3):231-237.
Domling et al., "Programmed Death-1: Therapeutic Success after More than 100 Years of Cancer Immunotherapy," Angew. Chem. Int. Ed., 2014, 53:2283-2288.
Dong et al., "Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion," Nature Medicine, Aug. 2002, 8(8):793-800.
Ecuador Opposition in Ecuador Application No. SENADI-2019-3773, dated Oct. 10, 2019, 29 pages.
Escarcega-Bobadilla et al., "A Recyclable Trinuclear Bifunctional Catalyst Derived from a Tetraoxo Bis-Zn(salphen) Metalloligand," Chemistry—A European Journal., 2013, 19(8):2641-2648.
Escarcega-Bobadilla et al., "Metal-directed assembly of chiral bis-Zn(II) Schiff base structures," Dalton Transactions, 2012, 41(32):9766-9772.
Escarcega-Bobadilla et al., "Versatile Switching in Substrate Topicity: Supramolecular Chirality Induction in Di- and Trinuclear Host Complexes," Chemistry—A European Journal, 2012:8(22):6805-6810.
Eurasian Office Action in Eurasian Application No. 201990074/28, dated Oct. 3, 2019, 5 pages.
European Communication in European Application No. 16805690.1, dated Jan. 22, 2020, 5 pages.
European Communication in European Application No. 16805690.1, dated Jul. 10, 2018, 6 pages.
European Communication in European Application No. 16805690.1, dated Nov. 5, 2020, 4 pages.
European Communication in European Application No. 17743174.9, dated Jan. 31, 2020, 5 pages.
European Communication in European Application No. 20202254.7, dated Apr. 1, 2022, 4 pages.
Fabris et al., "Central to Axial Transfer of Chirality in Menthone or Camphor-Derived 2,2′-Biphenols," J Org Chem., 1997, 62(21):7156-7164.
FDA Report, "22 Case Studies Where Phase 2 and Phase 3 Trials Had Divergent Results," U.S. Food and Drug Administration, Jan. 2017, 44 pages.
Francisco et al., "The PD-1 Pathway in Tolerance and Autoimmunity," Immunol. Rev., Jul. 2010, 236:219-242.
Freeman et al, "Engagement of the Pd-1 Immunoinhibitory Receptor by a Novel B7 Family Member Leads to Negative Regulation of Lymphocyte Activation," J. Exp. Med., Oct. 2000, 192(7):1027-34.
Freeman, "Structures of PD-1 with its ligands: Sideways and dancing cheek to cheek," PNAS, Jul. 2008, 105(30):10275-10276.
Freindorf, M., "Vibronic couplings in an excited state of hydrogen bond dimeric systems," Acta Physica Polonica, 1990, A78(6):825-839.
Gong et al., "Rhodium(I)-catalyzed regiospecific dimerization of aromatic acids: two direct C—H bond activations in water," Angewandte Chemie, 2015, 54(19):5718-5721.
Goswami et al., "A turn on ESIPT probe for rapid and ratiometric fluorogenic detection of homocysteine and cysteine in water with live cell-imaging," Tetrahedron Letters, 2014, 55(2):490-494.
Gould et al. "Salt selection for basic drugs," Int J Pharma., 1986, 33(1-3):201-217.
Green et al., "Synthesis and investigation of the configurational stability of some dimethylammonium borate salts," J. Chem. Soc., Perkin Trans. 1, 2000, 24:4403-4408.
Greenwald et al, "The B7 Family Revisited," Annu. Rev. Immunol., 2005, 23:515-548.
Gu et al., "Undo the brake of tumour immune tolerance with antibodies, peptide mimetics and small molecule compounds targeting PD-1/PD-L1 checkpoint at different locations for acceleration of cytotoxic immunity to cancer cells," Clinical and Experimental Pharmacology and Physiology, 2019, 46(2):105-115.
Han et al., "Synthesis of binuclear phenoxyimino organoaluminum complexes and their use as the catalyst precursors for efficient ring-opening polymerisation of E-caprolactone," Dalton Transactions, 2013, 41:12346-12353.
Helgeson et al., "Host-guest complexation. 66. 18-Membered-ring spherands containing five anisyl groups," J Am Chem Soc., 1993, 1115(24):11506-11511.
Highlights Prescribing Information, "KEYTRUDA," Revised Feb. 2019, 66 pages.
Highlights Prescribing Information, "OPDIVO," Revised Apr. 2019, 90 pages.
Hilfiker "Relevance of Solid-state Properties for Pharmaceutical Products," Polymorphism in the Pharmaceutical Industry, Jan. 1, 2006, pp. 1-19.
Hu et al., "Novel highly active binuclear neutral nickel and palladium complexes as precatalysts for norboernene polymerization," Journal of Molecular Catalysis A: Chemical 253, 2006, 155-164.
Hu et al., "Syntheses and Ethylene Polymerization Behavior of Supported Salicylaldimine-Based Neutral Nickel(II) Catalysts," Organometallics, 2007, 26(10):2609-2615.
Hu et al., "Synthesis and Ethylene Polymerization Activity of a Novel, Highly Active Single-Component Binuclear Neutral Nickel(II) Catalyst," Organometallics, 2005, 24(11):2628-2632.
Huang et al, "The prognostic significance of PD-L1 in bladder cancer," Oncol. Rep., 2015, 33:3075-3084.
Huang et al., "Pharmacological treatment for keloids," Expert opinion on pharmacotherapy, 2013, 14(15):2087-2100.
Huddle et al., "Reactions of alkyl-lithium compounds with aryl halides ," J Chem Soc., Perkin I, 1980, 12:2617-2625.
HuGEMM™ and HuCELL™ Models, "FactSheet," CrownBio, Oct. 2016, 8 pages.
Indian Office Action in Indian Application No. 202017053661, dated Jun. 3, 2022, 5 pages.
Indian Office Action with Indian Application No. 201817026809, dated Apr. 29, 2020, 6 pages.
Indian Office Action with Indian Application No. 201917001998, dated Nov. 24, 2020, 7 pages.
Indian Office Action with Indian Application No. 201917028273, dated Feb. 15, 2021, 5 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2016/057487, dated May 3, 2018, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2016/062730, dated May 31, 2018, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2016/067155, dated Jun. 19, 2018, 10 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2016/067925, dated Jun. 26, 2018, 8 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/031242, dated Nov. 6, 2018, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/034173, dated Nov. 27, 2018, 8 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/038120, dated Dec. 25, 2018, 6 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/041899, dated Jan. 15, 2019, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/048880, dated Mar. 5, 2019, 9 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/067880, dated Jun. 25, 2019, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/067886, dated Jun. 25, 2019, 9 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/067904, dated Jun. 25, 2019, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/067946, dated Jun. 25, 2019, 9 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/067951, dated Jun. 25, 2019, 8 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/067984, dated Jun. 25, 2019, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2019/025036, dated Oct. 15, 2020, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2019/031728, dated Nov. 17, 2020, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2020/045311, dated Feb. 17, 2022, 8 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2020/053190, dated Apr. 5, 2022, 7 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2020/059817, dated May 17, 2022, 12 pages.
International Search Report and Written Opinion in International Application No. PCT/US2016/057487, dated Dec. 8, 2016, 11 pages.
International Search Report and Written Opinion in International Application No. PCT/US2016/062730, dated Feb. 9, 2017, 12 pages.
International Search Report and Written Opinion in International Application No. PCT/US2016/067155, dated Apr. 24, 2017, 26 pages.
International Search Report and Written Opinion in International Application No. PCT/US2016/067925, dated Mar. 27, 2017, 13 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/031242, dated Jun. 20, 2017, 22 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/034173, dated Aug. 8, 2017, 15 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/038120, dated Aug. 1, 2017, 14 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/041899, dated Sep. 5, 2017, 12 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/048880, dated Oct. 23, 2017, 15 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/067880, dated Mar. 21, 2018, 13 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/067886, dated Mar. 23, 2018, 24 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/067904, dated Mar. 22, 2018, 14 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/067946, dated May 22, 2018, 16 Pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/067951, dated Mar. 27, 2018, 15 pages.
International Search Report and Written Opinion in International Application No. PCT/US2017/067984, dated Mar. 22, 2018, 14 pages.
International Search Report and Written Opinion in International Application No. PCT/US2019/025036, dated Jul. 3, 2019, 12 pages.
International Search Report and Written Opinion in International Application No. PCT/US2019/031728, dated Jun. 25, 2019, 12 pages.
International Search Report and Written Opinion in International Application No. PCT/US2020/045311, dated Oct. 2, 2020, 14 pages.
International Search Report and Written Opinion in International Application No. PCT/US2020/059817, dated Mar. 29, 2021, 19 pages.
International Search Report and Written Opinion in International Application No. PCT/US2021/058268, dated Apr. 21, 2022, 22 pages.
International Search Report and Written Opinion in International Application No. PCT/US2021/058334, dated Apr. 25, 2022, 23 pages.
International Search Report and Written Opinion in International Application No. PCT/US2021/058338, dated Feb. 9, 2022, 15 pages.
International Search Report and Written Opinion in International Application No. PCT/US2021/063965, dated Apr. 12, 2022, 20 pages.
International Search Report in International Application No. PCT/US2020/053190, dated Jan. 29, 2021, 13 pages.
Invitation to Pay Fee's in International Application No. PCT/US2021/058268, dated Jan. 31, 2022, 16 pages.
Invitation to Pay Fee's in International Application No. PCT/US2021/058334, dated Feb. 3, 2022, 12 pages.
Israeli Office Action in Israeli Application No. 259,406, dated Mar. 11, 2020, 10 pages.
Israeli Office Action in Israeli Application No. 260,166, dated Jun. 2, 2020, 13 pages.
Israeli Office Action in Israeli Application No. 287,267, dated Feb. 15, 2022, 4 pages.
Iwai et al, "Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade," PNAS, Sep. 2002, 99(19):12293-12297.
Japanese Office Action in Japanese Application No. 2018526213, dated Oct. 13, 2020, 10 pages.
Japanese Office Action in Japanese Application No. 2019-534122, dated Oct. 19, 2021, 10 pages.
Japanese Office Action in Japanese Application No. 2019-534195, dated Nov. 1, 2021, 9 pages.
Japanese Office Action in Japanese Application No. 2019-534196, dated Nov. 9, 2021, 9 pages.
Jiang et al., "Self-immobilizing binuclear neutral nickel catalyst for ethylene polymerization: Synthesis and catalytic studies," J Mol Cat., 2013, 380:139-143.
Kayal et al., "3,3′-Bis(triphenylsilyl)biphenoxide as a Sterically Hindered Ligand on Fe(II), Fe(III), and Cr(II)," Inorg Chem., 2002, 41(2):321-330.
Keir et al., "PD-1 and Its Ligands in Tolerance and Immunity," Annu. Rev. Immunol., 2008, 26:677-704.
Kerekes et al., "Aurora kinase inhibitors based on the imidazo[1,2-a]pyrazine core: fluorine and deuterium incorporation improve oral absorption and exposure," J Med Chem., 2011, 54(1):201-210.
Koch et al., "Nucleophilic reactions of pyridines and imidazoles with vinyl and aromatic halides," J Org Chem., 1993, 58(6):1409-1414.
Komiyama et al., "IL-17 Plays an Important Role in the Development of Experimental Autoimmune Encephalomyelitis," J. Immunol., Jul. 2006, 177:566-73.
Latchman et al, "PD-L2 is a second ligand for PD-1 and inhibits T cell activation," Nat. Immunol., Mar. 2001, 2(3):261-268.
Lazar-Molnar et al., "Crystal structure of the complex between programmed death-1 (PD-1) and its ligand PD-L2," PNAS, Jul. 2008, 105(30):10483-10488.
Legon'kova et al., "Interaction of o,o-dihalo o′-hydroxy azo compounds with metallic copper. II. Preparation of oligomeric azo compounds from monoazo compounds," Mosk Khim-Tekhnol Inst im Mendeleeva., 1968, 11(11):1281-1284 Machine Translation.
Legon'kova et al., "Interaction of o,o-dihalogeno o-hydroxy azo compounds with metallic copper," Trudy Instituta—Moskovskii Khimiko-Tekhnologicheskii Institut imeni D. I. Mendeleeva, 1965, 48:120-125 Machine Translation.
Lehtonen et al., "Comparison of quaternary methyl-, ethyl- and butylammonium hydroxides as alkylating reagents in pyrolysis-GC/MS studies of aquatic fulvic acid," Journal of Analytical and Applied Pyrolysis, 2003, 68-69:315-329.
Lexico.com, "Synonyms of Enhance," Oxford Dictionary, retrieved on Dec. 9, 2021, retrieved from URL <https://www.lexico.conn/synonynns/enhance>, 4 pages.
Li et al., "A 3D Mesomeric Supramolecular Structure of a Cu(II) Coordination Polymer with 1,1′-Biphenyl-2,2′,3,3′-tetracarboxylic Acid and 5,5′-Dimethyl-2,2′-bipyridine Ligands," J Inorg and Organomet Poly Mat., 2012, 22(6):1320-1324.
Li et al., "A Mini-Review for Cancer Immunotherapy: Molecular Understanding of PD-1/PD-L1 Pathway & Translational Blockade of Immune Checkpoints," Int. J. Mol. Soc., 2016, 17:1151, 22 pages.
Li et al., "Analysis of Receptor Tyrosine Kinase Internalization Using Flow Cytometry," Methods Mol. Biol., 2008, 457:305-317.
Li et al., "Asymmetric Alternating Copolymerization of Meso-epoxides and Cyclic Anhydrides: Efficient Access to Enantiopure Polyesters," J. Am. Chem. Soc., 2016, 138(36):11493-11496.
Li et al., "Discovery of peptide inhibitors targeting human programmed death 1 (PD-1) receptor," Oncotarget, Aug. 2016, 7(40):64967-64976.
Lin et al., "The PD-1/PD-L1 complex resembles the antigen-binding Fv domains of antibodies and T cell receptors," PNAS, Feb. 2008, 105(8):3011-3016.
Lipson et al., "From Discovery to Development: Blocking PD-1 and its Ligands," The Melanoma Letter, A Publication of The Skin Cancer Foundation, vol. 31, Summer 2013, 6 pages.
Liu et al., "Asymmetric Copolymerization of CO2 with meso-Epoxides Mediated by Dinuclear Cobalt(III) Complexes: Unprecedented Enantioselectivity and Activity," Angewandte Chemie, 2013, 52(44):11594-11598.
Liu et al., "Development of amino- and dimethylcarbamate-substituted resorcinol as programmed cell death-1 (PD-1) inhibitor," Eur J Pharm Sci, 2016, 88:50-58.
Mahoney et al., "The Next Immune-Checkpoint Inhibitors:PD-1/PD-L1 Blockade in Melanoma," Clin. Therapeutics, Nov. 2015, 37(4):761-782.
Maier et al., "Effects of the stationary phase and the solvent on the stereodynamics of biphep ligands quantified by dynamic three-column HPLC," Angewante Chemie, 2012, 51(12):2985-2988.
Manecke et al., "Preparation and properties of chelate-forming monomeric and polymeric Schiff bases derived from salicylaldehyde and 2,5-dihydroxyterephthalaldehyde. I," Makromolekulare Chemie, 1970, 133:61-82 English Abstract.
Manecke et al., "Preparation and properties of monomeric and polymeric Schiff bases derived from salicylaldehyde and 2,5-dihydroxyterephthalaldehyde. II. Electrical conductivity," Makromolekulare Chemie, 1972, 160:111-126 English Abstract.
Mexican Office Action in Mexican Application No. MX/a/2018/007774, dated Apr. 8, 2021, 5 pages.
Mexican Office Action in Mexican Application No. MX/a/2018/016273, dated Mar. 26, 2021, 5 pages.
Miyaura and Suzuki, "Palladium-Catalyzed Cross-Coupling Reactions of Organoboron Compounds," Chem Rev., 1995, 95:2457-2483.
Mochida et al., "Rhodium-Catalyzed Regioselective Olefination Directed by a Carboxylic Group," J Org Chem, 2011, 76(9):3024-3033.
Moneta et al., "Boron templated synthesis of macrocyclic hosts containing convergent hydroxy or methoxy groups," Bulletin de la Societe Chimique de France, 1988, 6:995-1004 (English Abstract).
Nallasivam et al., "Development of Unimolecular Tetrakis(piperidin-4-ol) as a Ligand for Suzuki-Miyaura Cross-Coupling Reactions: Synthesis of Incrustoporin and Preclamol," 2015, Eur J Org Chem., 2015(16):3558-3567.
Nero et al., "Oncogenic protein interfaces: small molecules, big challenges," Nature Reviews, Apr. 2014, 14:248-262.
Nishimura et al, "Autoimmune Dilated Cardiomyopathy in PD-1 Receptor-Deficient Mice," Science, Jan. 2001, 291:319-322.
Nishimura et al, "Development of Lupus-like Autoimmune Diseases by Disruption of the PD-1 Gene Encoding an ITIM Motif-Carrying Immunoreceptor," Immunity, Aug. 1999, 11:141-151.
Nishimura et al., "PD-1: an inhibitory immunoreceptor involved in peripheral tolerance," TRENDS in Immunology, May 2001, 22(5):365-268.
Nishino et al., "Copper-Mediated C—H/C—H Biaryl Coupling of Benzoic Acid Derivatives and 1,3-Azoles," Angew. Chem. Int. Ed., 2013, 52:4457-4461.
Normand et al., "Dinuclear vs. mononuclear complexes: accelerated, metal-dependent ring-opening polymerization of lactide," Chem. Commun., 2013, 49(99):11692-11694.
Okazaki and Honjo, "The PD-1-PD-L pathway in immunological tolerance," Trends Immunol., Apr. 2006, 4:195-201.
Okazaki et al., "A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application," Nature Immunology, Dec. 2013, 14(12):1212-1218.
Paek et al., "Chiral host. Attempted synthesis using McMurray reaction as a final ring closure method," Bulletin of the Korean Chemical Society, 1989, 10(6):572-577.
Paek et al.., "Facile syntheses and multi-orthofunctionalizations of tertiary benzamides," Bulletin of the Korean Chemical Society, 1993, 14(6):732-739.
Pardoll, "The blockade of immune checkpoints in cancer immunotherapy," Nature, Apr. 2012, 12:252-264.
Parry et al., "CTLA-4 and PD-1 Receptors Inhibit T-Cell Activation by Distinct Mechanisms," Mol. Cell. Biol., Nov. 2005, 25(21):9543-9553.
Parsons et al., "Directed ortho metalation reactions. Expedient synthesis of 3,3′-disubstituted 1,1′-bi-(2-phenols) (BIPOLS)," Tetrahedron Letters, 1994, 35(41):7537-7540.
Pascolutti et al., "Structure and Dynamics of PD-L1 and an Ultra-High-Affinity PD-1 Receptor Mutant," Structure, Oct. 2016, 24:1719-1728.
Paulini et al., "Orthogonal Multipolar Interactions in Structural Chemistry and Biology," Angew. Chem. Int. Ed., 2005, 44:1788-1805.
Pearson et al., "The formation of complexes between aza-derivatives of crown ethers and primary alkylammonium salts. Part 5. Chiral macrocyclic diamines," J. Chem. Soc., Perkin I, 1979, 12:3113-3126.
Pfeiffer et al., "Inner complex salts of the aldimine and azo series," Journal fuer Praktische Chemie, 1937, 149:217-296 Machine Translation.
Pierre et al., "Synthesis of a new macrobicyclic siderophoric host molecule with six converging phenolate groups," Angewandte Chemie, 1991, 103(1):75-76 Machine Translation.
Postow et al, "Immune Checkpoint Blockade in Cancer Therapy," J. Clinical Oncology, Jun. 2015, 33(17):1974-1982.
Press Release Archive, "Boehringer Ingelheim and Yale University collaborate to investigate novel immunotherapy targets across several therapeutic areas," Boehringer Ingelheim, Jan. 13, 2015, 2 pages.
Puehlhofer et al., "SASAPOS cascades of perfluorinated aromatic carboxylic acids: low-temperature decarboxylation triggered by electrostatic effects of polycationic ligand sets," Euro J of Org Chem., 2004, 5:1002-1007.
Punniyamurthy et al., "Enantiomerically pure bicyclo[3.3.1]nona-2,6-diene as the sole source of enantioselectivity in BIPHEP-Rh asymmetric hydrogenation," Chem Comm., 2008, 41:5092-5094.
Qin et al., "The Diverse Function of PD-1/PD-L Pathway Beyond Cancer," Frontiers In Imunology, Oct. 2019, 10(2298):1-16.
Reck et al., "Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer," N Engl J Med., Nov. 10, 2016, 375(19):1823-1833.
Rowe et al., "Fumaric Acid" Handbook of pharmaceutical excipients, Jan. 1, 2009, pp. 276-277, 309-310, 393-396.
Sabatier et al, "Prognostic and predictive value of PDL1 expression in breast cancer," Oncotarget, Mar. 2015, 6(7):5449-5464.
Sharma et al., "Palladium-Catalyzed Decarboxylative Acylation of O-Phenyl Carbamates with Alpha-Oxocarboxylic Acids at Room Temperature," Advanced Synthesis & Catalysis, 2013, 355(4):667-672.
Sharpe et al, "The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection," Nat. Immunol., Mar. 2007 8(3):239-245.
Sharpe et al., "The B7-CD28 Superfamily," Nature Reviews, Feb. 2002, 2:116-126.
Sorrell et al., "3,3′-Disubstituted 2,2′-biphenols. Synthesis of nonplanar, tetradentate chelating ligands," J Org Chem., 1985, 50(26):5765-5769.
Stahl et al., Handbook of Pharmaceutical Salts: Properties, Selection, and Use, (Wiley, 2002), p. 329-350. *
STN Search Report dated Apr. 14, 2016, 79 pages.
STN Search Report dated Apr. 29, 2016, 69 pages.
STN Search Report dated Apr. 30, 2018, 8 pages.
STN Search Report dated Aug. 19, 2016, 23 pages.
STN Search Report dated Aug. 30, 2016, 4 pages.
STN Search Report dated Dec. 15, 2016, 4 pages.
STN Search Report dated Dec. 16, 2016, 25 pages.
STN Search Report dated Dec. 16, 2016, 4 pages.
STN Search Report dated Dec. 19, 2016, 11 pages.
STN Search Report dated Dec. 20, 2016, 117 pages.
STN Search Report dated Jul. 12, 2016, 4 pages.
STN Search Report dated Jun. 16, 2016, 8 pages.
STN Search Report dated Jun. 6, 2016, 115 pages.
STN Search Report dated Mar. 27, 2018, 4 pages.
STN Search Report dated May 24, 2016, 92 pages.
STN Search Report dated Sep. 12, 2016, 17 pages.
STN Search Report dated Sep. 12, 2016, 4 pages.
STN Search Report dated Sep. 2, 2016, 115 pages.
STN Search Report dated Sep. 27, 2017, 4 pages.
STN Search Report, dated May 1, 2016, 12 pages.
Storz, "Intellectual property issues of immune checkpoint inhibitors," mAbs, Jan. 2016, 8(1):10-26.
Suarez et al., "Inhibitors of TAM subfamily of tyrosine kinases: synthesis and biological evaluation," European Journal of Medicinal Chemistry, 2013, 61:2-25.
Sumrit et al., "Aluminum complexes containing salicylbenzoxazole ligands and their application in the ring-opening polymerization of rac-lactide and ϵ-caprolactone," Dalton Transactions (2016), 45(22), 9250-9266.
Sun et al., "Studies on Synthesis and Properties of Some New Dibenzocyclobromonium," Chemical Journal of Chinese Universities, 1998, 19(12), 6 pages (English Abstract).
Sznol et al., "Antagonist Antibodies to PD-1 and B7-H1 (PD-L1) in the Treatment of Advanced Human Cancer," Clin Cancer Res., Mar. 1, 2013, 19(5):1021-1034.
Taiwan Office Action in Taiwan Application No. 105133530, dated Oct. 15, 2020, 8 pages.
Taiwan Office Action in Taiwan Application No. 105137807, dated Nov. 12, 2020, 12 pages.
Taiwan Office Action in Taiwan Application No. 105141804, dated Nov. 9, 2020, 9 pages.
Tang et al., "Facile synthesis of enantioenriched phenol-sulfoxides and their aluminum complexes," Org Biomol Chem., 2016, 14(24):5580-5585.
Thiel et al., "Small-Molecule Stabilization of Protein-Protein Interactions: An Underestimated Concept in Dmg Discoveiy?" Angew. Chem. Int. Ed., 2012, 51:2012-2018.
Tucker et al., "Host-guest complexation. 52. Bridged and chiral hemispherands," J Org Chem., 1989, 54(23):5460-5482.
Ukraine Office Action in Ukraine Application No. a 2019 00525, dated Jan. 14, 2021, 11 pages.
Unrau et al., "Directed ortho metalation. Suzuki cross coupling connections. Convenient regiospecific routes to functionalized m- and p-teraryls and m-quinquearyls," Tetrahedron Letters, 1992, 33(20):2773-2776.
Vaddepally et al., "Review of Indications of FDA-Approved Immune Checkpoint Inhibitors per NCCN Guidelines with the Level of Evidence," Cancers, 2020, 12(3):738.
Velcheti et al., "Programmed death-1/programmed death-1 ligand axis as a therapeutic target in oncology: current insights," Journal of Receptor Ligand and Channel Research, Dec. 2014, 8(23): 1-7.
Wang et al, "The prognostic value of PD-L1 expression for non-small cell lung cancer patients: A meta-analysis," Eur. J. Surg. Oncol., 2015, 41:450-456.
Wang et al., "A binuclear Zn(II)-Zn(II) complex from a 2-hydroxybenzohydrazide-derived Schiff base for selective detection of pyrophosphate," Dalton Transactions, Oct. 2014, 43(37):14142-14146.
Wang et al., "Molecular Modeling and Functional Mapping of B7-H1 and B7-DC Uncouple Costimulatory Function from PD-1 Interaction," J. Exp. Med., Apr. 2013, 197(3):1083-1091.
Wei et al., "Strength of PD-1 signaling differentially affects T-cell effector functions," PNAS, Apr. 2013, E2480-E2489.
Weinmann, "Cancer Immunotherapy: Selected Targets and Small-Molecule Modulators," Chem. Med. Chem., 2016, 11:450-466.
Weiss et al., "Electrostatic activation of SNAr-reactivity by sulfonylonio substituents," Zeitschrift fuer Naturforschung, 2001, 56(12): 1360-1368 English Abstract.
Weiss et al., "Electrostatics and color: Massive electrostatic perturbation of chromophores by ion cluster ligands," J Am Chem Soc., 2007, 129(3):547-553.
Weiss et al., "First-ever per(onio) substitution of benzene: the role of the counterion," Angewandte Chemie, 1995, 34(12):1319-1321.
Weiss et al., "Massive electrostatic effects on heteropolar C—C disconnections: Transforming a phenyl anion into a potent leaving group," Euro J Org Chem., 2005, 16:3530-3535.
Weiss et al., "Poly-onio substituted quinones as strong electron acceptors," Inst Org Chem., 1986, 98(10):925-926.
Weiss et al., "SASAPOS, not Sisyphus: highly efficient 20-step one-pot synthesis of a discrete organic-inorganic ion cluster with a porphyrin core," Angewandte Chemie International Edition, 2002, 41(20):3815-3817.
Weiss et al., "Syntheses and Reactions of Polycationically Substituted Azido- and Diazidobenzenes," Eur J Org Chem., Nov. 2007, 31:5270-5276.
Wells et al., "Reaching for high-hanging fruit in dmg discovery at protein-protein interfaces," Nature, Dec. 2007, 450:1001-1009.
Wu et al., "Targeting the BACE1 Active Site Flap Leads to a Potent Inhibitor That Elicits Robust Brain Aβ Reduction in Rodents," ACS Medicinal Chemistry Letters, 2016, 7(3):271-276.
Wuts et al., "Protective Groups in Organic Synthesis," 4th Ed., 2007, 1111 pages.
www.medscape.com' [online]. "The ‘Family Business’ Behind the Flurry of PD-1 Inhibitors," Sep. 10, 2014. [Retrieved on Jan. 29, 2015]. Retrieved from the Internet: URL<http://www.medscape.com/viewarticle/831448_print>. 3 pages.
Xiong et al., "Biaryl-Bridged Salalen Ligands and Their Application in Titanium-Catalyzed Asymmetric Epoxidation of Olefins with Aqueous H2O2," Eur J Org Chem., 2011, 23:4289-4292.
Xu et al., "Design, synthesis and biological evaluation of deuterated nintedanib for improving pharmacokinetic properties," J Label Compd RadioPharm., Jun. 15, 2015, 58(7):308-312.
Xu et al., "Quantitative structure-activity relationship study on BTK inhibitors by modified multivariate adaptive regression spline and CoMSIA methods," SAR QSAR Environ Res., 2015, 26(4):279-300.
Yao et al., "PD-1 as an Immune Modulatory Receptor," Cancer J., 2014, 20(4):262-264.
Yin et al., "Strategies for Targeting Protein-Protein Interactions With Synthetic Agents," Angew. Chem. Int. Ed., 2005, 44:4130-4163.
Young et al., "Discovery of highly potent and selective Bruton's tyrosine kinase inhibitors: Pyridazinone analogs with improved metabolic stability," Bioorganic & Medicinal Chemistry Letters, 2016, 26(2):575-579.
Young et al., "Potent and selective Bruton's tyrosine kinase inhibitors: Discovery of GDC-0834," Bioorganic & Medicinal Chemistry Letters, 2015, 25(6):1333-1337.
Zak et al., "Structural basis for small molecule targeting of the programmed death ligand 1 (PD-L1)," Oncotarget, 2016, 7(21):30323-30335.
Zak et al., "Structural basis for small molecule targeting of the programmed death ligand 1 (PD-L1)" Oncotarget, Apr. 2016, 19 pages; Supplemental Material for 2016, 7(21):30323-30335.
Zak et al., "Structure of the Complex of Human Programmed Death 1, PD-1, and Its Ligand PD-L1: with Supplemental Information," Structure, Dec. 2015, 23:2341-2348.
Zang et al., "Four 2D metal-organic networks incorporating Cd-cluster SUBs: hydrothermal synthesis, structures and photoluminescent properties," CrystEngComm, 2009, 11(1):122-129.
Zarganes-Tzitzikas, "Inhibitors of programmed cell death 1 (PD-1): a patent review (2010-2015)," Expert Opinion on Therapeutic Patents, Sep. 19, 2016, 26(9):973-977.
Zhan et al., "From monoclonal antibodies to small molecules: the development of inhibitors targeting the PD-1/PD-L1 pathway," Drug Discovery Today, Apr. 2016, 10 pages.
Zhang et al., "Biaryl-Based Macrocyclic and Polymeric Chiral (Salophen)Ni(II) Complexes: Synthesis and Spectroscopic Study," J Org Chem., 2001, 66(2):481-487.
Zhang et al., "Electrospray mass spectrum of a per(onio)-substituted benzene: retention of Coulombic charge upon collisionally activated decomposition," J Am Soc. Mass. Spectrom., 1998, 9(1):15-20.
Zhang et al., "Non-symmetrical diarylcarboxylic acids via rhodium(I)-catalyzed regiospecific cross-dehydrogenation coupling of aromatic acids: twofold direct C—H bond activations in water," RSC Advances, 2016, 6(64):91617-91620.
Zhang et al., "Structural and Functional Analysis of the Costimulatory Receptor Programmed Death-1," Immunity, Mar. 2004, 20:337-347.
Zhao et al., "Design, synthesis and organocatalysis of 2,2′-biphenol-based prolinamide organocatalysts in the asymmetric direct aldol reaction in water," Synlett, 2013, 24(20):2743-2747.

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11866435B2 (en) 2015-12-22 2024-01-09 Incyte Corporation Heterocyclic compounds as immunomodulators

Also Published As

Publication number Publication date
TW202115059A (zh) 2021-04-16
MX2022001671A (es) 2022-05-13
JP2022544189A (ja) 2022-10-17
WO2021030162A1 (fr) 2021-02-18
US20210040090A1 (en) 2021-02-11
AR119624A1 (es) 2021-12-29
CA3150434A1 (fr) 2021-02-18
EP4010342A1 (fr) 2022-06-15

Similar Documents

Publication Publication Date Title
US11753406B2 (en) Salts of a PD-1/PD-L1 inhibitor
US11760756B2 (en) Crystalline form of a PD-1/PD-L1 inhibitor
US11718605B2 (en) Heterocyclic compounds as immunomodulators
US11866451B2 (en) Salts and crystalline forms of a PD-1/PD-L1 inhibitor
US11613536B2 (en) Heterocyclic compounds as immunomodulators
EP3774791B1 (fr) Composés hétérocycliques utilisés comme immunomodulateurs
US11401279B2 (en) Pyrido[3,2-d]pyrimidine compounds as immunomodulators
EP3558989B1 (fr) Dérivés de triazolo[1,5-a]pyridine en tant qu&#39;immunomodulateurs
EP3558973B1 (fr) Dérivés pyridine utilisés en tant qu&#39;immunomodulateurs
IL302590A (en) Process for preparing PD-1/PD-L1 inhibitor and salts and crystalline forms thereof
CN116514715A (zh) 作为免疫调节剂的杂环化合物

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

AS Assignment

Owner name: INCYTE CORPORATION, DELAWARE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JIA, ZHONGJIANG;LIU, PINGLI;MELONI, DAVID J.;AND OTHERS;SIGNING DATES FROM 20201104 TO 20201109;REEL/FRAME:055727/0139

Owner name: WILMINGTON PHARMATECH, DELAWARE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LI, QUN;REEL/FRAME:055727/0245

Effective date: 20201026

Owner name: INCYTE CORPORATION, DELAWARE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WILMINGTON PHARMATECH;REEL/FRAME:055727/0269

Effective date: 20201111

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE